Next Article in Journal
New Genetic Markers of Skin T-Cell Lymphoma Treatment
Previous Article in Journal
Variation of FMRP Expression in Peripheral Blood Mononuclear Cells from Individuals with Fragile X Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Whole-Exome Sequencing (WES) Reveals Novel Sex-Specific Gene Variants in Non-Alcoholic Steatohepatitis (MASH)

Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University Health Sciences, Spokane, WA 99202, USA
*
Author to whom correspondence should be addressed.
Genes 2024, 15(3), 357; https://doi.org/10.3390/genes15030357
Submission received: 13 February 2024 / Revised: 6 March 2024 / Accepted: 11 March 2024 / Published: 13 March 2024

Abstract

:
Non-alcoholic steatohepatitis (NASH, also known as MASH) is a severe form of non-alcoholic fatty liver disease (NAFLD, also known as MASLD). Emerging data indicate that the progression of the disease to MASH is higher in postmenopausal women and that genetic susceptibility increases the risk of MASH-related cirrhosis. This study aimed to investigate the association between genetic polymorphisms in MASH and sexual dimorphism. We applied whole-exome sequencing (WES) to identify gene variants in 8 age-adjusted matched pairs of livers from both male and female patients. Sequencing alignment, variant calling, and annotation were performed using standard methods. Polymerase chain reaction (PCR) coupled with Sanger sequencing and immunoblot analysis were used to validate specific gene variants. cBioPortal and Gene Set Enrichment Analysis (GSEA) were used for actionable target analysis. We identified 148,881 gene variants, representing 57,121 and 50,150 variants in the female and male cohorts, respectively, of which 251 were highly significant and MASH sex-specific (p < 0.0286). Polymorphisms in CAPN14, SLC37A3, BAZ1A, SRP54, MYH11, ABCC1, and RNFT1 were highly expressed in male liver samples. In female samples, Polymorphisms in RGSL1, SLC17A2, HFE, NLRC5, ACTN4, SBF1, and ALPK2 were identified. A heterozygous variant 1151G>T located on 18q21.32 for ALPK2 (rs3809983) was validated by Sanger sequencing and expressed only in female samples. Immunoblot analysis confirmed that the protein level of β-catenin in female samples was 2-fold higher than normal, whereas ALPK2 expression was 0.5-fold lower than normal. No changes in the protein levels of either ALPK2 or β-catenin were observed in male samples. Our study suggests that the perturbation of canonical Wnt/β-catenin signaling observed in postmenopausal women with MASH could be the result of polymorphisms in ALPK2.

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) (also known as metabolic dysfunction-associated fatty liver disease, MAFLD) is the leading cause of chronic liver disease, affecting 25% of the US population [1,2]. It is commonly associated with obesity, diabetes, and metabolic syndrome but can also affect non-obese individuals. The disease spectrum ranges from bland steatosis with or without inflammation (non-alcoholic fatty liver, NAFL) to steatosis with inflammation and hepatocellular injury (non-alcoholic steatohepatitis, NASH) (also known as metabolic dysfunction-associated steatotic liver disease, MASH), fibrosis, cirrhosis, and hepatocellular carcinoma [3]. Owing to the lack of reliable noninvasive predictive biomarkers, the diagnosis of MASH is mainly limited to the histopathological evaluation of liver samples defined by liver-biopsy-proven hepatocellular steatosis, lobular inflammation, and evidence of hepatocyte injury such as ballooning degeneration [4]. A large body of evidence strongly supports the idea that MASLD susceptibility and progression to MASH are sex specific. Several studies conducted in single centers or in specific populations have suggested that women have a 19% lower risk of MASLD than men in the general population. However, once MASLD has become established, women have a 37% higher risk of advanced fibrosis than men [5]. Among individuals with established MASLD who are older than 50 years, women have a 17% greater risk for MASH and a 56% greater risk for advanced fibrosis than men [5,6]. Although it has been established that the prevalence of risk factors such as age, obesity, type 2 diabetes mellitus (T2DM), atherogenic dyslipidemia, and clinical outcomes of MASLD differs between sexes, the molecular mechanisms by which sex modulates the pathogenesis and clinical outcomes of MASLD progression are poorly defined. Therefore, to understand the potential mechanisms underlying this sexual dimorphism in MASLD prevalence, we recently used a multiomics approach with archived liver samples from both sexes to study the biological basis of the observed sexual dimorphism. Our study suggests (for the first time) that the activation of canonical Wnt signaling could be one of the main pathways associated with sexual dimorphism in MASLD and MASH [7].
Two different Wnt signaling pathways, canonical and non-canonical, have their own influence on MASLD and MASH. The non-canonical pathway is involved in the accumulation of fat, inflammation, and lipids, which promote MASH formation. The canonical pathway involving β-catenin functions as an anti-inflammatory, anti-lipid accretion, and adipocyte differentiation pathway [8]. Hence, the inhibition or downregulation of the classical Wnt/β-catenin pathway contributes to the onset and progression of MASLD. For example, MASLD is inhibited by the upregulation of peroxisome proliferator activated receptor γ (PPAR-γ), a downstream target of the Wnt/β-catenin signaling that promotes preadipocyte differentiation, adipogenesis, the absorption of free fatty acids (FFA), and the suppression of inflammation [9]. Polymorphisms in low-density lipoprotein receptor-related protein-6 (LRP6) are a major cause of MASLD [10]. Although it is well documented that MASLD progression is attributed to dynamic interactions between genetic and environmental factors [11], there is still limited information on how canonical Wnt/β-catenin signaling is involved in MASLD/MASH disease progression. Therefore, we hypothesized that gene variants in the Wnt/β-catenin signaling pathway could be associated with the observed sexual dimorphism in MASH, as suggested by our recent study [7].
To test this hypothesis, we used whole-exome sequencing (WES) to identify potential gene variants implicated in MASH using 16 archived frozen liver samples from paired males and females. Here, we report the identification of α protein kinase 2 (ALPK2) gene variants (rs3809981and rs3809983) as female-specific single-nucleotide polymorphisms (SNPs) in the postmenopausal livers of women with MASH.

2. Methods

2.1. Ethics Statement

The Institutional Review Board (IRB) of Washington State University (WSU) approved the protocol of the current study. Sixteen paired matched snap-frozen tissue samples were obtained from the IRB-approved University of Minnesota Liver Tissue Cell Distribution System (LTCDS). All specimens with anonymized identifiers were histopathologically confirmed by a pathologist (Table S1; Supplemental Digital Content).

2.2. DNA Extraction and Whole-Exome Sequencing (WES-Seq)

Genomic DNA was extracted from 16 frozen liver tissue samples (4 matched pairs of both sexes) using a Wizard Genomic DNA purification kit (A1120, Promega, Madison, WI, USA) following the manufacturer’s instructions. The DNA concentration was measured using a NanoDrop spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA). The extracted DNA (50 ng/uL/sample) was shipped to LC Sciences (Houston, TX, USA) for exome sequencing (100× coverage). Two hundred nanograms of genomic DNA (200 ng) from each subject’s MASH-normal paired samples, which were fragmented by sonication, were subjected to library preparation using the Agilent SureSelect Human All Exon V6 kit (Agilent Technologies, Santa Clara, CA, USA) following the vendor’s recommended protocol. DNA libraries were hybridized and captured using SureSelect. Following hybridization, the captured libraries were purified according to the manufacturer’s instructions and amplified by polymerase chain reaction (PCR). Normalized libraries were pooled, and DNA was subjected to paired-end sequencing using the Illumina HiSeq X Ten platform with a 150-bp paired-end sequencing mode.

2.3. WES Data Processing

Raw sequence reads were trimmed to remove low-quality sequences and then aligned to the human reference genome (hg19) using the Burrows–Wheeler alignment tool [12]. Single-nucleotide polymorphisms and small insertions/deletions were identified in individual samples using the Genome Analysis Toolkit (GATK Mutect2 4.0.4.0) with the default setting [13]. ANNOVAR was then used to annotate the VCF files using the gene region and several filters from other databases [14]. Finally, we used the Database for Annotation, Visualization, and Integrated Discovery (DAVID) Bioinformatics Resource 6.7 (https://David-d.ncifcrf.gov, accessed on 22 July 2023) and Gene Set Enrichment Analysis (GESA) [15] to identify significantly altered biological processes and pathways in 16 liver tissue samples.

2.4. PCR and Sanger Sequencing

To validate the ALPK2 polymorphisms, we used PCR and Sanger sequencing from Azenta Life Sciences (Burlington, MA, USA). Specific PCR primers for ALPK2, F: TGCTGTCTATCAAATCTCGGCT and R: GAGCACTCAACCTCAACGGA were used. Primers were designed using Primer3 (http://bioinfo.ut.ee/primer3-0.4.0/, accessed on 22 July 2023). The products were directly sequenced using the ABI PRISM BigDye Kit on an ABI 3130 DNA sequencer (Applied Biosystems, Foster City, CA, USA). Sequencing results were analyzed using A Plasmid Editor [16].

2.5. Western Blot Analysis

Frozen liver tissue samples (n = 12) were homogenized in ice-cold lysis buffer containing a protease/phosphatase inhibitor cocktail and centrifuged at 12,000× g at 4 °C for 15 min. Protein samples were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto polyvinylidene difluoride (PVDF) membranes. After blocking in 5% non-fat milk at 37 °C for 1 h, the membranes were incubated overnight at 4 °C with primary antibodies against ALPK2 (ab111909, Abcam, Cambridge, UK), β-catenin (8480S, Cell Signaling Technology, Danvers, MA, USA), or GAPDH (sc-47724, Santa Cruz Biotechnology, Dallas, TX, USA). Following incubation with the secondary antibody, immunoreactive proteins were visualized using the ChemiDoc Touch Imaging System (Bio-Rad). Protein bands were quantified using the ImageJ 1.53k.

2.6. Statistical Analysis of Western Blot

The data were expressed as the mean ± SEM (n = 3/phenotype/sex) and Student’s t-test was used to analyze statistical significance. Statistical analyses were performed, and graphs were generated using GraphPad Prism 6 (GraphPad Software Inc., San Diego, CA, USA). ** p < 0.01 was considered statistically significant.

3. Results

3.1. Clinical Characteristics of the Study Population

Sixteen snap-frozen liver tissue samples (normal and MASH) from white non-Hispanic populations of both sexes were used in this study. The median age (range) of patients was 54 to 59 years old. In general, the clinicopathological characteristics of patients with MASH (steatosis, steatohepatitis, ballooning, and portal inflammation) were higher in women than in men. Detailed clinicopathological information is summarized in Supplemental Table S1.

3.2. WES, Data Filtering and Mutation Landscape of Liver Tissue Samples

As shown in Figure 1, using the WES approach we identified 148,881 gene variants in 16 liver tissue samples, representing 57,121 and 50,150 gene variants in female and male cohorts, respectively. For SNVs, 35,000 (27%) were exonic and 79,259 (59%) were intronic (Table 1). For InDels, 13,925 were identified and 10,837 (78%) were intronic, as shown in Table 1. Our analysis detected no differences in SNPs, InDel distribution, or mutation type between sexes (Supplemental Figures S1 and S2). By contrast, FACETS analysis [17] revealed that copy number variants (CNVs) in female cohorts differed from those in male cohorts. As shown in Figure 2A, many gene variants (female cases), such as SLC17A2 (Table 2), were clustered around chromosome 6 (as represented by allele-specific log-odd-ratio data), whereas in male cases (Figure 2B), many gene variants such as CAPN14 (Table 3) were clustered around chromosome 11. Collectively, these observations suggest that copy-number alterations (CNAs) of these genes are different in the two cohorts and could play an important role in the sexual dimorphism of MASH.

3.3. WES Identifies ALPK2 Variant in Female Cases

To further analyze our gene variant data, statistical significance was first determined via a hypothetical Fisher’s exact test (Figure 1); with four male samples vs. four females, a polymorphism was considered significant if it existed in all female samples but none of the male samples (or if a polymorphism existed in all male samples but none of the female samples). The corresponding p-value for this assumption was 0.0286. We merged and filtered the vcf files of individual samples and searched for polymorphisms that met the above criteria. Polymorphisms that passed the criteria were then annotated with the Var2GO tool [18], using GRCh37 as a reference, given that the original analysis was performed using the hg19 genome. A total of 251 highly significant sex-specific MASH gene variants (p < 0.0286) were identified. A total of 63 MASH female-specific gene variants were identified, as shown in Table 2, whereas 54 gene variants were identified in males (Table 3). Among the 54 male variants, we found polymorphisms in CPN14 (12 intronic variants), SRP54 (four intronic and upstream gene variants), ABCC1 (three synonymous and intronic gene variants), RNFT1 (two upstream gene variants), SLC37A3 (two intronic and upstream gene variants), obg-like ATPase 1 (OLA1) (two intronic and non-synonymous variants), BAZ1A (two intronic and downstream gene variants), and MYH11 (two intronic and synonymous variants).
Of the 63 female variants (Table 2), we identified SCL17A (six intronic and synonymous variants), RGSL1 (three intronic variants), ACTN4 (three synonymous and upstream gene variants), NLRC5 (two synonymous and non-synonymous variants), BIN1 (two intronic variants), C7 (two intronic and downstream gene variants), HIST1H4B (synonymous and downstream gene variants), SBF1 (two upstream gene variants), and ALPK2 (two synonymous and non-synonymous variants). In this study, we validated α Protein Kinase 2 (ALPK2) as a novel genetic variant associated with MASH in a female cohort.

3.4. Validation of the ALPK2 Variant

To identify the biological pathways associated with ALPK2, we performed gene set enrichment analysis (GSEA) [15] using a TCGA liver cancer patient cohort from the cBioPortal database. As shown in Table 4, Wnt gene signatures, including canonical/β-catenin-mediated pathways, were negatively enriched in ALPK2-high (FDR q-val = 0.036 to 0.003) vs. ALPK2-low samples (FDR q-val = 0.105 to 0.544), which is consistent with a previous report showing ALPK2 as a negative regulator of canonical Wnt signaling [19]. These data also confirmed that ALPK2 is associated with β-catenin-mediated pathways in women with MASH, as we previously reported [7].
Next, we validated the ALPK2 mutation by PCR testing coupled with Sanger sequencing. As shown in Figure 3, the normal, healthy sample HH1202 was used as a reference for comparison with the two female MASH samples (UMN1535 and UMN1259). A clear single nucleotide polymorphism (SNP) is highlighted with a black box in the MASH samples in Figure 3A,B. The identified SNP (p.Ala1551Ser) resulted in nsSNV (rs3809983), as shown in Table 2.
Since ALPK2 was shown to be involved in the canonical Wnt/β-catenin signaling pathway (Table 4), we measured the protein expression of both ALPK2 and β-catenin in both male and female liver tissue samples using immunoblot analysis. As shown in Figure 4A,B, the protein expression of β-catenin in female samples was 2-fold higher than that in normal samples, whereas ALPK2 expression was 0.5-fold lower than that in normal samples. No change in the expression of either ALPK2 or β-catenin was observed in male samples (Figure 4C,D).

4. Discussion

Genetics play a key role in MASLD pathogenesis [20,21]. Variations in genes such as patatin-like phospholipase domain-containing protein 3 (PNPLA3), transmembrane 6 superfamily member 2 (TM6SF2), membrane-bound O-acyltransferase domain-containing 7 (MBOAT7), glucokinase regulator (GCKR), and hydroxysteroid 17-β dehydrogenase-13 (HSD17B13) have emerged as reproducibly and robustly predisposing individuals to the development of MASH [22,23]. However, despite these discoveries, some unexplained variance remains, indicating that additional genetic associations with MASLD/MASH may be revealed using multi-omics analyses.
Although sex differences exist in the prevalence, risk factors, fibrosis, and clinical outcomes of MASLD/MASH, our understanding of the genetic basis of sexual dimorphism remains limited. Therefore, in this study, we performed WES analyses of paired-matched liver tissue samples from male and female MASH patients (Table S1) to elucidate sex-specific gene variants associated with this disease. As shown in Figure 1, we identified 63 gene variants that were specific to the female and 54 male-specific variants (Fisher’s exact test p < 0.0286). Interestingly, a significant number of these gene variants have been identified with respect to the sexual dimorphism of MASLD/MASH, whereas others have been previously reported to be involved in the pathogenesis of the disease. For example, in male-specific variants (Table 3), we identified CAPN14 as encoding a calcium-regulated non-lysosomal thiol-protease (Calpain) as a top gene variant that is known to be involved in a variety of cellular processes including apoptosis, cell division, the modulation of integrin–cytoskeletal interactions, and synaptic plasticity [24]. Recently, calpains have been shown to be associated with hepatocyte death in MASH and the progression of hepatocellular carcinoma (HCC) [25,26]. Regarding chr2 (2p23.1), we found that OLA1 encodes a member of the GTPase protein family. It interacts with breast-cancer-associated gene 1 (BRCA1) and BRCA1-associated RING domain protein (BRAD1) and is involved in centrosome regulation [27]. OLA1 has been shown to be associated with hereditary breast and ovarian cancers as well as with a poor prognosis of HCC [28,29]. Polymorphisms were also found in other canonical cancer-related genes, including SLC37A3, BAZ1A, SRP54, MYH1 and ABCC1 [30,31,32,33], but were not directly involved in MASLD pathogenesis. As shown in Table 3, we also identified a SNP (synonymous variant) in ORAI1 (ORAI calcium release-activated calcium modulator 1), which encodes a membrane calcium channel subunit activated by the calcium sensor STIM1 when calcium stores are depleted [34]. ORAI polymorphisms have been shown to be associated with non-canonical Wnt signaling, MASLD progression, and HCC [35,36].
For female-specific gene variants (Table 2), we identified six loci of SLC17A2 on chr6 (6p22.2), encoding proteins belonging to sodium-dependent phosphate transporters. A recent study reported that SLC17A2 variants were associated with MASLD in lean individuals [37]. In the present study, SLC17A2 was specifically identified in female MASH patients. For the same chr6 (6p22.2), we also established that HFE encodes a transmembrane protein that regulates iron absorption by regulating the interaction of the transferrin receptor with transferrin associated with MASLD in lean individuals along with SLC17A2 (37). For chr16 (16q13), we identified two loci NLRC5 that encode members of the caspase recruitment domain of the NLR family. This gene plays a major role in the regulation of the NF-kappa B and interferon signaling pathways [38]. Polymorphisms in NLRC5 are associated with obesity, type 2 diabetes mellitus (T2DM), and MASLD [39] and limit the NF-kB signaling pathway [40].
In the present study, we identified rs3809983 ALPK2 as a novel gene variant associated with MASH in female liver samples. ALPK2 mapped to 18q21.32 encodes a serine/threonine kinase protein that is involved in several processes, including epicardium morphogenesis and heart development, and is a negative regulator of Wnt signaling [19]. Recent studies by McIntosh et al. [41] showed that ALPK2 rs3809973 (not ALPK2 rs3809983, identified in this study) is associated with an increased risk of liver fibrosis in HIV/HCV co-infected women. This may be the initial indication linking the ALPK2 variant to the pathological liver phenotype in women. Furthermore, Lawrence et al. [42] found that ALPK2 is a novel polymorphic gene in human cancers in a large-scale genomic analysis of 4742 human neoplasms and their matched normal tissue samples. In mouse xenograft models, the knockdown of ALPK2 inhibits the development and progression of ovarian cancer [43] and renal cancer cells [44], thus supporting its relevance not only in cancer initiation and development but also in the pathogenesis of liver disease.
To validate the ALPK2 polymorphism, we used PCR coupled with Sanger sequencing and found that ALPK2 rs3809983 was associated with MASH in the female patient samples (Figure 3). This association was further confirmed by immunoblot analysis (Figure 4), suggesting that the ALPK2 polymorphism was linked to defective canonical Wnt signal transduction only in female samples. ALPK2 polymorphisms cause inappropriate levels of β-catenin and thus a perturbation of the Wnt signaling pathway in female patients with MASH, as we previously reported [7]. These observations thus agree with the cBioPortal analysis (Table 4), suggesting a good correlation between ALPK2 loss/decreased function and the loss of its negative regulatory activity in the canonical Wnt/β-catenin signaling pathway.
Despite the important findings of this study, it has some limitations. These limitations are primarily associated with the availability of paired matched MASLD/MASH liver samples from the male and female cohorts. Although the present study was limited by the relatively small number of available samples, the data presented here showed a clear and robust distinction between female and male patients with respect to gene variants associated with MASH livers compared with normal livers. We hypothesize that future efforts should be made to increase the sample size while improving the selection of extreme phenotypes to maximize the power of this strategy. Demographic variables such as ethnic background should be considered in future studies. Owing to sample availability, the individuals included in our study were mainly of Caucasian origin, which may limit the applicability of our findings to other ethnic populations. These limitations highlight the critical need to improve research in this area, especially in clinically relevant conditions associated with MASLD and MASH such as inter-hepatic cholangiocarcinoma and celiac disease [45,46]. Further studies are also needed to elucidate the cellular and molecular basis on how ALPK2 variants may impact the sexual dimorphism of MASLD/MASH disease progression.
In summary, this study provides evidence that MASLD-related sexual dimorphism is influenced by genetic variants. We used WES of the liver tissue samples to identify sex-specific gene polymorphisms associated with MASH. Our study further provides evidence that polymorphisms in ALPK2 are associated with postmenopausal women compared to men and that the activation of the canonical Wnt signaling pathway previously reported [7] could be the result of ALPK2 polymorphisms. Other (downstream) members of the Wnt signaling pathway could also be associated with MASH severity in postmenopausal women compared to men.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes15030357/s1.

Author Contributions

Conception and design of the experiments: S.S.D. and B.J.W. Performing the experiments: J.W. Analysis of the data: J.W. and S.S.D. Contributing reagents/materials/analysis tools: B.J.W. and S.S.D. Writing of the paper: S.S.D. and B.J.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by CPPS-WSU (17A-2957-9838) to SSD, and NIH/NCI grants R37CA233658 and R01CA258634, and CPPS-WSU start-up funding to BJW.

Institutional Review Board Statement

The Washington State University (WSU) Office of Research Assurances has found that the study is exempt from the need for Institutional Research Board (IRB) approval. Sixteen snap-frozen tissue samples were obtained from the IRB-approved University of Minnesota Liver Tissue Cell Distribution System (Minneapolis, MN). All specimens with anonymized identifiers were histopathologically confirmed by a pathologist.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data supporting the findings of this study are available from the corresponding author upon reasonable request. The data are not publicly available due to privacy.

Acknowledgments

Human liver specimens were obtained using the Liver Tissue Cell Distribution System (Minneapolis, MN), which is funded by the National Institute of Health Contract No. N01-DK-7-0004/HHSN267200700004C. The authors would like to thank Phillip Wibisono for his valuable input in sequencing data analysis.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

NAFLD: non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis; MASLD, metabolic dysfunction-associated fatty liver disease; MASH, metabolic dysfunction-associated fatty liver disease; WES, whole-exome sequencing; SNP, single nucleotide polymorphism; nsSNV, non-synonymous single nucleotide variant; InDels, insertions and deletions; HCC, hepatocellular carcinoma; ALPK2, α kinase 2; GSEA, gene set enrichment analysis; FACETS, fraction and allele-specific copy number estimates from tumor sequencing; PCR, polymerase chain rection.

References

  1. Younossi, Z.; Anstee, Q.M.; Marietti, M.; Hardy, T.; Henry, L.; Eslam, M.; George, J.; Bugianesi, E. Global burden of NAFLD and NASH: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 11–20. [Google Scholar] [CrossRef]
  2. Chalasani, N.; Younossi, Z.; LaVine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the study of liver diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef]
  3. Araujo, A.R.; Rosso, N.; Bedogni, G.; Tiribelli, C.; Bellentani, S. Global epidemiology of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis: What we are need in the future. Liver Int. 2017, 38, 47–51. [Google Scholar] [CrossRef] [PubMed]
  4. Koch, L.K.; Yeh, M.M. Nonalcoholic fatty liver disease (NAFLD): Diagnosis, pitfalls, and staging. Ann. Diagn. Pathol. 2018, 37, 83–90. [Google Scholar] [CrossRef] [PubMed]
  5. Balakrishnan, M.; Patel, P.; Dunn-Valadez, S.; Dao, C.; Khan, V.; Ali, H.; El-Serag, L.; Hernaez, R.; Sisson, A.; Thrift, A.P.; et al. Women have a lower risk of nonalcoholic fatty liver disease but higher risk of progression vs men: A systematic review and meta-analysis. Clin. Gastroenterol. Hepatol. 2021, 19, 61–71. [Google Scholar] [CrossRef]
  6. Burra, P.; Bizzaro, D.; Gonta, A.; Shalaby, S.; Gambato, M.; Morelli, M.C.; Trapani, S.; Floreani, A.; Marra, F.; Brunetto, M.R.; et al. Clinical impact of sexual dimorphism in non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Liver Int. 2021, 41, 1713–1733. [Google Scholar] [CrossRef]
  7. Yeh, M.M.; Shi, X.; Yang, J.; Li, M.; Fung, K.M.; Daoud, S.S. Perturbation of Wnt/β-catenin signaling and sexual dimorphism in non-alcoholic fatty liver disease. Hepatol. Res. 2022, 52, 433–448. [Google Scholar] [CrossRef] [PubMed]
  8. Ackers, I.; Malgor, R. Interrelationship of canonical and non-canonical Wnt signaling pathways in chronic metabolic diseases. Diabetes Vasc. Dis. Res. 2018, 15, 3–13. [Google Scholar] [CrossRef]
  9. Wang, X.M.; Wang, X.Y.; Huang, Y.M.; Chen, X.; Lü, M.H.; Shi, L.; Li, C.P. Role and mechanisms of action of microRNA21 as regards the regulation of the Wnt/β-catenin signaling pathway in the pathogenesis of non-alcoholic fatty liver disease. Int. J. Mol. Med. 2019, 44, 2201–2212. [Google Scholar]
  10. Go, G.W. Low-density lipoprotein receptor-related 6 (LRP6) is a novel nutritional therapeutic target for hyperlipidemia, non-alcoholic fatty liver disease, and atherosclerosis. Nutrients 2015, 7, 4453–4464. [Google Scholar] [CrossRef]
  11. Albhaisi, S.; Sanyal, A. Gene-environmental interactions as metabolic drivers of nonalcoholic steatohepatitis. Front. Endocrinol. 2021, 12, 665987. [Google Scholar] [CrossRef] [PubMed]
  12. Büchler, T.; Ohlebusch, E. An improved encoding of genetic variations in a Burrows-Wheeler transform. Bioinformatics 2020, 36, 1413–1419. [Google Scholar] [CrossRef] [PubMed]
  13. Ulintz, P.J.; Wu, W.; Gates, C.M. Bioinformatics analysis of whole exome sequencing data. Methods Mol. Biol. 2019, 1881, 277–318. [Google Scholar] [PubMed]
  14. Yang, H.; Wang, K. Genomic variant annotation, and prioritization with ANNOVAR and wANNOVAR. Nat. Protoc. 2015, 10, 1556–1566. [Google Scholar] [CrossRef]
  15. Zito, A.; Lualdi, M.; Granata, P.; Cocciadiferro, D.; Novelli, A.; Alberio, T.; Casalone, R.; Fasano, M. Gene set enrichment analysis of interaction networks weighted by node centrality. Front. Genet. 2021, 12, 577623. [Google Scholar] [CrossRef]
  16. Davis, M.W.; Jorgensen, E.M. ApE, A Plasmid Editor: A freely available DNA manipulation and visualization program. Front. Bioinform. 2022, 2, 818619. [Google Scholar] [CrossRef] [PubMed]
  17. Shen, R.; Seshan, V.E. FACETS: Allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 2016, 44, e131. [Google Scholar] [CrossRef]
  18. Granata, I.; Sangiovanni, M.; Maiorano, F.; Miele, M.; Guarracino, M.R. Var2GO: A web-based tool for gene variants selection. BMC Bioinform. 2016, 11, 376–382. [Google Scholar] [CrossRef]
  19. Hofsteen, P.; Robitaille, A.M.; Strash, N.; Palpant, N.; Moon, R.T.; Pabon, L.; Murry, C.E. ALPK2 promotes cardiogenesis in Zebrafish and human pluripotent stem cells. iScience 2018, 2, 88–100. [Google Scholar] [CrossRef]
  20. Eslam, M.; George, J. Genetic contribution to NAFLD: Leveraging shared genetics to uncover system biology. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 40–52. [Google Scholar] [CrossRef]
  21. Sookoian, S.; Pirola, C.J. Genetic predisposition in nonalcoholic fatty liver disease. Clin. Mol. Hepatol. 2017, 23, 1–12. [Google Scholar] [CrossRef] [PubMed]
  22. Luukkonen, P.K.; Qadri, S.; Ahlholm, N.; Porthan, K.; Männistö, V.; Sammalkorpi, H.; Penttilä, A.K.; Hakkarainen, A.; Lehtimäki, T.E.; Gaggini, M.; et al. Distinct contributions of metabolic dysfunction and genetic risk factors in the pathogenesis of non-alcoholic fatty liver disease. J. Hepatol. 2022, 76, 526–535. [Google Scholar] [CrossRef] [PubMed]
  23. Carlsson, B.; Lindén, D.; Brolén, G.; Liljeblad, M.; Bjursell, M.; Romeo, S.; Loomba, R. The emerging role of genetics in precision medicine for patients with non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 2020, 51, 1305–1320. [Google Scholar] [CrossRef]
  24. Ma, X.-L.; Zhu, K.-Y.; Chen, Y.-D.; Tang, W.-G.; Xie, S.-H.; Zheng, H.; Tong, Y.; Wang, Y.-C.; Ren, N.; Guo, L.; et al. Identification of a novel Calpain-2-SRC feed-back loop as necessity for β-Catenin accumulation and signaling activation in hepatocellular carcinoma. Oncogene 2022, 41, 3554–3569. [Google Scholar] [CrossRef] [PubMed]
  25. Seike, T.; Boontem, P.; Yanagi, M.; Li, S.; Kido, H.; Yamamiya, D.; Nakagawa, H.; Okada, H.; Yamashita, T.; Harada, K.; et al. Hydroxynonenal causes hepatocyte death by disrupting lysosomal integrity in non-alcoholic steatohepatitis. Cell. Mol. Gastroenterol. Hepatol. 2022, 14, 925–944. [Google Scholar] [CrossRef] [PubMed]
  26. Dai, D.; Wu, D.; Ni, R.; Li, P.; Tian, Z.; Shui, Y.; Hu, H.; Wei, Q. Novel insights into the progression and prognosis of the calpain family members in hepatocellular carcinoma: A comprehensive integrated analysis. Front. Mol. Biosci. 2023, 10, 1162409–1162422. [Google Scholar] [CrossRef]
  27. Matsuzawa, A.; Kanno, S.I.; Nakayama, M.; Mochiduki, H.; Wei, L.; Shimaoka, T.; Furukawa, Y.; Kato, K.; Shibata, S.; Yasui, A.; et al. The BRCA1/BARD1-interacting protein OLA1 functions in centrosome regulation. Mol. Cell. 2014, 53, 101–114. [Google Scholar] [CrossRef]
  28. Takahashi, M.; Chiba, N.; Shimodaira, H.; Yoshino, Y.; Mori, T.; Sumii, M.; Nomizu, T.; Ishioka, C. OLA1 gene sequencing in patients with BRCA1/2 mutation negative suspected hereditary breast and ovarian cancer. Breast Cancer 2017, 24, 336–340. [Google Scholar] [CrossRef]
  29. Huang, S.; Zhang, C.; Sun, C.; Hou, Y.; Zhang, Y.; Tam, N.L.; Wang, Z.; Yu, J.; Huang, B.; Zhuang, H.; et al. Obg-like ATPase 1 (OLA1) overexpression predicts poor prognosis and promotes tumor progression by regulating P21/CDK2 in hepatocellular carcinoma. Aging 2020, 12, 3025–3041. [Google Scholar] [CrossRef]
  30. Meng, Z.; Geng, X.; Lin, X.; Wang, Z.; Chen, D.; Liang, H.; Zhu, Y.; Sui, Y. A prospective diagnostic and prognostic biomarker for hepatocellular carcinoma that functions in glucose metabolism regulation: Solute carrier family 37 member 3. Biochim. Biophys. Acta Mol. Basis Dis. 2023, 1868, 166661. [Google Scholar] [CrossRef]
  31. Millo, T.; Rivera, A.; Obolensky, A.; Marks-Ohana, D.; Xu, M.; Li, Y.; Wilhelm, E.; Gopalakrishnan, P.; Gross, M.; Rosin, B.; et al. Identification of autosomal recessive novel genes and retinal phenotypes in members of the solute carrier (SLC) superfamily. Genet. Med. 2022, 24, 1523–1535. [Google Scholar] [CrossRef] [PubMed]
  32. Ishikawa, Y.; Kawashima, N.; Atsuta, Y.; Sugiura, I.; Sawa, M.; Dobashi, N.; Yokoyama, H.; Doki, N.; Tomita, A.; Kiguchi, T.; et al. Prospective evaluation of prognostic impact of KIT mutations on acute myeloid leukemia with RUNX1-RUNX1T1 and CBFB-MYH11. Blood Adv. 2020, 4, 66–75. [Google Scholar] [CrossRef] [PubMed]
  33. Kadioglu, O.; Saeed, M.; Munder, M.; Spuller, A.; Greten, H.J.; Efferth, T. Identification of novel ABCC1 transporter mutations in tumor biopsies of cancer patients. Cells 2020, 9, 299. [Google Scholar] [CrossRef] [PubMed]
  34. Michelucci, A.; Garcia-Castaneda, M.; Boncompagni, S.; Kirksen, R.T. Role of STIM1/ORAI1-mediated store-operated Ca2+ entry in skeletal muscle physiology and disease. Cell Calcium 2018, 76, 101–115. [Google Scholar] [CrossRef] [PubMed]
  35. Ali, E.S.; Rychkov, G.; Barritt, G. Metabolic disorders, and cancer: Hepatocyte store operated Ca2+ channels in nonalcoholic fatty liver disease. Adv. Exp. Med. Biol. 2017, 993, 595–621. [Google Scholar]
  36. Petko, J.; Thileepan, M.; Sargen, M.; Canfield, V.; Levenson, R. Alternative splicing of the Wnt trafficking protein, Wntless and its effects on protein-protein interactions. BMC Mol. Cell Biol. 2019, 20, 22–31. [Google Scholar] [CrossRef]
  37. Sun, Z.; Pan, X.; Tian, A.; Surakka, I.; Wang, T.; Jiao, X.; He, S.; Song, J.; Tian, X.; Tong, D.; et al. Genetic variants in HFE are associated with non-alcoholic fatty liver disease in lean individuals. JHEP Rep. 2023, 5, 100744. [Google Scholar] [CrossRef]
  38. Li, Y.Y.; Chung, G.T.; Lui, V.W.; To, K.F.; Ma, B.B.; Chow, C.; Woo, J.K.S.; Yip, K.Y.; Seo, J.; Hui, E.P.; et al. Exome and genome sequencing of nasopharynx cancer identifies NF-kB pathway activating mutations. Nat. Commun. 2017, 8, 14121–14131. [Google Scholar] [CrossRef]
  39. Bauer, S.; Hezinger, L.; Rexhepi, F.; Ramanathan, S.; Kufer, T.A. NOD-like receptors-emerging links to obesity and associated morbidities. Int. J. Mol. Sci. 2023, 24, 8595. [Google Scholar] [CrossRef]
  40. Chang, G.; Liu, X.; Ma, T.; Xu, L.; Wang, H.; Li, Z.; Guo, X.; Xu, Q.; Chen, G. A mutation in the NLRC5 promoter limits NF-kB signaling after Salmonella Enteritidis infection in the spleen of young chickens. Gene 2015, 568, 117–123. [Google Scholar] [CrossRef] [PubMed]
  41. McIntosh, A.T.; Wei, R.; Ahn, J.; Aouizerat, B.E.; Kassaye, S.G.; Augenbraun, M.H.; Price, J.C.; French, A.L.; Gange, S.J.; Anastos, K.M.; et al. A genomic variant of ALPK2 is associated with increased liver fibrosis risk in HIV/HCV coinfected women. PLoS ONE 2021, 16, e0247277. [Google Scholar] [CrossRef] [PubMed]
  42. Lawrence, M.S.; Stojanov, P.; Mermel, C.H.; Robinson, J.T.; Garraway, L.A.; Golub, T.R.; Meyerson, M.; Gabriel, S.B.; Lander, E.S.; Getz, G. Discovery and saturation analysis of cancer genes across 21 tumor types. Nature 2014, 505, 495–501. [Google Scholar] [CrossRef] [PubMed]
  43. Zhu, X.; Yan, S.; Xiao, S.; Xue, M. Knockdown of ALPK2 inhibits the development and progression of ovarian cancer. Cancer Cell Int. 2020, 20, 267–277. [Google Scholar] [CrossRef] [PubMed]
  44. Jiang, J.; Han, P.; Qian, J.; Zhang, S.; Wang, S.; Cao, Q.; Shao, P. Knockdown of ALPK2 blocks development and progression of renal cell carcinoma. Exp. Cell Res. 2020, 392, 112029. [Google Scholar] [CrossRef]
  45. De Lorenzo, S.; Tovoli, F.; Mazzotta, A.; Vasuri, F.; Edeline, J.; Malvi, D.; Boudjema, K.; Renzulli, M.; Jeddou, H.; D’errico, A.; et al. Non-alcoholic steatohepatitis as a risk factor for interhepatic cholangiocarcinoma and its prognostic role. Cancers 2020, 12, 3182. [Google Scholar] [CrossRef]
  46. Tovoli, F.; Negrini, G.; Farì, R.; Guidetti, E.; Faggiano, C.; Napoli, L.; Bolondi, L.; Granito, A. Increased risk of nonalcoholic fatty liver disease in patients with coelic disease on a gluten-free diet: Beyond traditional metabolic factor. Aliment. Pharmacol. Ther. 2018, 48, 538–546. [Google Scholar] [CrossRef]
Figure 1. Illustration of WES workflow from frozen liver tissue samples of male and female patients to MASH sex-specific gene variants. Pipeline of bioinformatics analysis adapted in the WES results of gene variants.
Figure 1. Illustration of WES workflow from frozen liver tissue samples of male and female patients to MASH sex-specific gene variants. Pipeline of bioinformatics analysis adapted in the WES results of gene variants.
Genes 15 00357 g001
Figure 2. A representative integrated visualization of FACETS analysis of WES data for (A) female and (B) male total copy number variants (CNVs). The top panel displays total copy number log-ratio (logR), and the second panel displays allele-specific log-odds-ratio data (logOR) with chromosomes alternating in blue and gray. The third panel plots the corresponding integer (total, minor) copy number calls. The overall ploidy and purity for female patients in this case are 2.03 and 0.65, respectively, and 2.05 and 0.63 for male patients. The estimated cellular fraction (cf) profile is plotted at the bottom, revealing the aggregate of variants at each chromosome.
Figure 2. A representative integrated visualization of FACETS analysis of WES data for (A) female and (B) male total copy number variants (CNVs). The top panel displays total copy number log-ratio (logR), and the second panel displays allele-specific log-odds-ratio data (logOR) with chromosomes alternating in blue and gray. The third panel plots the corresponding integer (total, minor) copy number calls. The overall ploidy and purity for female patients in this case are 2.03 and 0.65, respectively, and 2.05 and 0.63 for male patients. The estimated cellular fraction (cf) profile is plotted at the bottom, revealing the aggregate of variants at each chromosome.
Genes 15 00357 g002aGenes 15 00357 g002b
Figure 3. Representative of Sanger sequence alignment (A) and chromatograms (B) of ALPK2 in normal and MASH female livers. Sequencing alignment was performed using a plasmid editor. A normal representative liver sample with no SNPs (HH1202) was used as a reference for comparison against two MASH-related samples (UMN1535 and UMN1259). A clear SNP is highlighted with a black box. The SNP leads to a substitution mutation from a hydrophobic alanine (A) at the 1151 position to a polar serine (S). No SNPs were observed in MASH-related samples of male patients.
Figure 3. Representative of Sanger sequence alignment (A) and chromatograms (B) of ALPK2 in normal and MASH female livers. Sequencing alignment was performed using a plasmid editor. A normal representative liver sample with no SNPs (HH1202) was used as a reference for comparison against two MASH-related samples (UMN1535 and UMN1259). A clear SNP is highlighted with a black box. The SNP leads to a substitution mutation from a hydrophobic alanine (A) at the 1151 position to a polar serine (S). No SNPs were observed in MASH-related samples of male patients.
Genes 15 00357 g003aGenes 15 00357 g003b
Figure 4. Immunoblot analysis of ALPK2 and β-catenin in female (A,B) and male (C,D) liver tissue samples. ALPK2 and β-catenin protein band intensity results were normalized to GAPDH and quantitatively analyzed with ImageJ 1.53k.. The ratio of target protein to GAPDH in individual normal groups was set as 1. Data represent the mean ± SEM. ** p < 0.01; ns, not significant; n = 3 samples/phenotype/sex.
Figure 4. Immunoblot analysis of ALPK2 and β-catenin in female (A,B) and male (C,D) liver tissue samples. ALPK2 and β-catenin protein band intensity results were normalized to GAPDH and quantitatively analyzed with ImageJ 1.53k.. The ratio of target protein to GAPDH in individual normal groups was set as 1. Data represent the mean ± SEM. ** p < 0.01; ns, not significant; n = 3 samples/phenotype/sex.
Genes 15 00357 g004
Table 1. Statistics of somatic SNV and InDels in position.
Table 1. Statistics of somatic SNV and InDels in position.
TypeSNVInDels
Counts
Percent
Counts
Percent
Downstream580
0.4
72
0.5
Exonic35,000
27
733
5
Exonic; splicing17
0
4
0
Intergenic3941
3
371
3
Intronic 79,259
59
10,837
78
ncRNA_exonic3011
2
252
2
ncRNA_intronic4444
3
550
4
ncRNA_splicing8
0
0
0
Splicing111
0.1
47
0.3
Upstream1228
1
115
1
Upstream; downstream115
0.1
5
0
UTR34504
3
687
5
UTR52730
2
247
2
UTR5; UTR313
0
5
0
All134,961
100
13,925
100
SNV = s ingle nucleotide variant; InDels = insertion/deletion mutations.
Table 2. Female common uniquely significant annotated variants.
Table 2. Female common uniquely significant annotated variants.
#CHR
OM
POSGene_IDGene_NameCytoBandAvsnp150CategoryREFALTGene_Full_Namep-Value
chr117085589ENSG00000186715MST1L1p36.13rs3863807upstream_gene_variantAGC
GCTG
Amacrophage stimulating 1-like0.0286
chr126487940ENSG00000197245FAM110D1p36.11rs3748856missense_variantAGfamily with sequence similarity 110 member D0.0286
chr126496455ENSG00000142684ZNF5931p36.11rs22326485_prime_UTR_premature
_start_codon_gain_variant
CTzinc finger protein 5930.0286
chr1154941593ENSG00000160691SHC11q21.3rs4845401upstream_gene_variantCGSHC (Src homology 2 domain containing) transforming protein 10.0286
chr1182509292ENSG00000121446RGSL11q25.3rs266531intron_variantAGregulator of G-protein signaling like 10.0286
chr1182509617ENSG00000121446RGSL11q25.3rs3911280intron_variantCAregulator of G-protein signaling like 10.0286
chr1182517357ENSG00000121446RGSL11q25.3rs6657620intron_variantGCregulator of G-protein signaling like 10.0286
chr1232172374ENSG00000162946DISC11q42.2rs17773715intron_variantGATSNAX-DISC1 readthrough (NMD candidate)0.0286
chr255176112ENSG00000214595EML62p16.1rs13394146intron_variantCTechinoderm microtubule associated protein like 60.0286
chr284668155ENSG00000163541SUCLG12p11.2rs115384987downstream_gene_variantTCsuccinate-CoA ligase, α subunit0.0286
chr2127808226ENSG00000136717BIN12q14.3rs2071270intron_variantATbridging integrator 10.0286
chr2127821085ENSG00000136717BIN12q14.3rs2071268intron_variantCTbridging integrator 10.0286
chr2202526366ENSG00000082126MPP42q33.1rs62193397downstream_gene_variantGAmembrane protein, palmitoylated 40.0286
chr332933360ENSG00000206557TRIM713p22.3rs3727941413_prime_UTR_variantCT,CTTtripartite motif containing 71, E3 ubiquitin protein ligase0.0286
chr357431721ENSG00000559559DNAH123p14.3rs372891308missense_variantAAAATAdynein axonemal heavy chain 120.0286
chr4110896050ENSG00000138798EGF4q25rs2067004sequence_featureACepidermal growth factor0.0286
chr540980086ENSG00000112936C75p13.1rs1450664splice_region_variant
and intron_variant
TCcomplement component 70.0286
chr540981689ENSG00000112936C75p13.1rs10614293_prime_UTR_variantCAcomplement component 70.0286
chr625914801ENSG00000112337SLC17A26p22.2rs62394272missense_variantGAsolute carrier family 17 member 20.0286
chr625914901ENSG00000112337SLC17A26p22.2rs2071298splice_region_variant
and intron_variant
GAsolute carrier family 17 member 20.0286
chr625916979ENSG00000112337SLC17A26p22.2rs1865760synonymous_variantCTsolute carrier family 17 member 20.0286
chr625918688ENSG00000112337SLC17A26p22.2rs1865760intron_variantGAsolute carrier family 17 member 20.0286
chr625924158ENSG00000112337SLC17A26p22.2rs1540273intron_variantTCsolute carrier family 17 member 20.0286
chr625925823ENSG00000112337SLC17A26p22.2rs7770139intron_variantAGsolute carrier family 17 member 20.0286
chr626027135ENSG00000124529HIST1H4B6p22.2rs37524203_prime_UTR_variantGAhistone cluster 1, H4b0.0286
chr626027433ENSG00000124529HIST1H4B6p22.2rs3752419synonymous_variantGAhistone cluster 1, H4b0.0286
chr626087856ENSG00000010704HFE6p22.2rs2858993intron_variantTAhomeostatic iron regulator0.0286
chr671011831ENSG00000112280COL9A16q13rs2242589intron_variantCTcollagen type IX α 10.0286
chr699819556ENSG00000132423COQ36q16.2rs4574651downstream_gene_variantCTcoenzyme Q3 methyltransferase0.0286
chr6152679729ENSG00000131018SYNE16q25.2rs9478326intron_variantGAspectrin repeat containing nuclear envelope 10.0286
chr7142498813ENSG00000211772TRBC27q34rs1042955synonymous_variantGAT cell receptor β constant 20.0286
chr8103301555ENSG00000104517UBR58q22.3rs2168689intron_variantTCubiquitin protein ligase E3 component n-recognin 50.0286
chr9107593182ENSG00000165029ABCA19q31.1rs4743763intron_variantATATP binding cassette subfamily A member 10.0286
chr1047701275ENSG00000198250ANTXRL10q11.22rs10906952synonymous SNVGAanthrax toxin receptor-like0.0286
chr10126480381ENSG00000203791METTL1010q26.13rs965484missense_variantCTEEF1A lysine methyltransferase 20.0286
chr1172309540ENSG00000186642PDE2A11q13.4rs4943939upstream_gene_variantCTphosphodiesterase 2A0.0286
chr129750669ENSG00000111796KLRB112p13.31rs1135816nonsynonymous SNVAGkiller cell lectin like receptor B10.0286
chr1253880122ENSG00000139625MAP3K1212q13.13rs3816806upstream_gene_variantTCmitogen-activated protein kinase 120.0286
chr1253896984ENSG00000139546TARBP212q13.13rs22804483_prime_UTR_variantGATAR (HIV-1) RNA binding protein 20.0286
chr1256865338ENSG00000135423GLS212q13.3rs2657879nonsynonymous SNVAGglutaminase 20.0286
chr1256866334ENSG00000135517MIP12q13.3rs2657880upstream_gene_variantTAmajor intrinsic factor of lens fiber0.0286
chr1288448328ENSG00000133641C12orf2912q21.32rs17418744downstream_gene_variantTAcentrosomal protein 290kDa0.0286
chr12119419632ENSG00000139767SRRM412q24.23rs15689245_prime_UTR_variantCTserine/arginine repetitive matrix 40.0286
chr1465414976ENSG00000139998RAB1514q23.3rs115408713_prime_UTR_variantCTRAB15, member RAS oncogene family0.0286
chr1471215822ENSG00000006432MAP3K914q24.2rs79518608downstream_gene_variantTCmitogen-activated protein kinase 90.0286
chr14105268104ENSG00000179627ZBTB4214q32.33rs10141867synonymous_variantGAzinc finger and BTB domain containing 420.0286
chr14107211211ENSG00000211976IGHV3-7314q32.33rs2073668synonymous_variantGAimmunoglobulin heavy variable 3-730.0286
chr1657075379ENSG00000140853NLRC516q13rs35622257missense_variantGGTNLR family, CARD domain containing 50.0286
chr1657080528ENSG00000140853NLRC516q13rs289723nonsynonymous SNVCANLR family, CARD domain containing 50.0286
chr1712832063ENSG00000006740ARHGAP4417p12rs1317990intron_variantGTRho GTPase activating protein 440.0286
chr1776867017ENSG00000035862TIMP217q25.3rs2277698synonymous_variantCTTIMP metallopeptidase inhibitor 20.0286
chr1856202768ENSG00000198796ALPK218q21.32rs3809983nonsynonymous SNVCAα kinase 20.0286
chr1856203120ENSG00000198796ALPK218q21.32rs3809981synonymous_variantCTα kinase 20.0286
chr1877724726ENSG00000226742HSBP1L118q23rs80957645_prime_UTR_variantACheat shock factor binding protein 1-like 10.0286
chr1917091368ENSG00000160111CPAMD819p13.11rs8103646synonymous_variantTGC3- and PZP-like, α-2-macroglobulin domain containing 80.0286
chr1939138608ENSG00000130402ACTN419q13.2rs2303040upstream_gene_variantTCactinin α 40.0286
chr1939196745ENSG00000130402ACTN419q13.2rs3745859synonymous SNVCTactinin α 40.0286
chr1939215333ENSG00000130402ACTN419q13.2rs3786851upstream_gene_variantCTactinin α 40.0286
chr1955644442ENSG00000105048TNNT119q13.42rs891186downstream_gene_variantGAtroponin T1, slow skeletal type0.0286
chr201617069ENSG00000089012SIRPG20p13rs2277761synonymous_variantAGsignal regulatory protein γ0.0286
chr2229834766ENSG00000128250RFPL122q12.2rs4657365_prime_UTR_variantAGRFPL1 antisense RNA 10.0286
chr2250906518ENSG00000100241SBF122q13.33rs1983679upstream_gene_variantGASET binding factor 10.0286
chr2250906917ENSG00000100241SBF122q13.33rs9616852upstream_gene_variantCASET binding factor 10.0286
chrX149937404ENSG00000102181CD99L2Xq28rs413116903_prime_UTR_variantTCCD99 molecule-like 20.0286
Table 3. Male common uniquely significant annotated variants.
Table 3. Male common uniquely significant annotated variants.
#CHR
OM
POSGene_IDGene_NameCytoBandAvsnp150CategoryREFALTGene_Full_Namep-Value
chr1114515717ENSG00000163349HIPK11p13.2rs2358996synonymous_variantGAhomeodomain interacting
protein kinase 1
0.0286
chr1234573357ENSG00000059588TARBP11q42.2rs2273875intron_variantGCTAR (HIV-1) RNA binding protein 10.0286
chr1237817784ENSG00000198626RYR21q43rs669375intron_variantAGryanodine receptor 20.0286
chr231397696ENSG00000214711CAPN142p23.1rs10180369intron_variantGCcalpain 140.0286
chr231397727ENSG00000214711CAPN142p23.1rs10180369intron_variantTCcalpain 140.0286
chr231399659ENSG00000214711CAPN142p23.1rs6720151intron_variantTCcalpain 140.0286
chr231399751ENSG00000214711CAPN142p23.1rs6720254intron_variantTGcalpain 140.0286
chr231399988ENSG00000214711CAPN142p23.1rs4592896non-synonymous SNVCTcalpain 140.0286
chr231400039ENSG00000214711CAPN142p23.1rs4516476intron_variantAGcalpain 140.0286
chr231400502ENSG00000214711CAPN142p23.1rs13421721intron_variantACcalpain 140.0286
chr231400510ENSG00000214711CAPN142p23.1rs1443707intron_variantGAcalpain 140.0286
chr231400722ENSG00000214711CAPN142p23.1rs1443706intron_variantGAcalpain 140.0286
chr231400867ENSG00000214711CAPN142p23.1rs1373216intron_variantTCcalpain 140.0286
chr231401499ENSG00000214711CAPN142p23.1rs28684727intron_variantGAcalpain 140.0286
chr231403947ENSG00000214711CAPN142p23.1rs2028678intron_variantGAcalpain 140.0286
chr2174946760ENSG00000138430OLA12q31.1rs11558990non-synonymous SNVTCObg-like ATPase 10.0286
chr2174988189ENSG00000138430OLA12q31.1rs10930639intron_variantCTObg-like ATPase 10.0286
chr2175199895ENSG00000231453AC018470.42q31.1rs3856434downstream_gene_variantGASp9 transcription factor0.0286
chr342772038ENSG00000244607CCDC133p22.1rs12495805non-synonymous SNVATcoiled-coil domain containing 130.0286
chr3124646594ENSG00000173702MUC133q21.2rs4679394non-synonymous SNVAGmucin 13, cell-surface-associated0.0286
chr3190967779ENSG00000188729OSTN3q28rs2034771intron_variantAGosteocrin0.0286
chr491645179ENSG00000184305CCSER14q22.1rs62314447intron_variantATmultimerin 10.0286
chr647253631ENSG00000146072TNFRSF216p12.3rs11758366intron_variantAGtumor necrosis factor receptor
superfamily member 21
0.0286
chr73861353ENSG00000146555SDK17p22.2rs6943646intron_variantCGsidekick cell adhesion molecule 10.0286
chr772396170ENSG00000196313POM1217q11.23rs782134793intron_variantGCGCCGCG
CTCCCCAC
GPOM121 transmembrane nucleoporin0.0286
chr7140036999ENSG00000157800SLC37A37q34rs4332050intron_variantGAsolute carrier family 37 member 30.0286
chr7140044979ENSG00000157800SLC37A37q34rs6974016upstream_gene_variantCTsolute carrier family 37 member 30.0286
chr9100889340ENSG00000106789CORO2A9q22.33rs942165intron_variantGTcoronin 2A0.0286
chr1051549314ENSG00000138294MSMB10q11.23rs12770171upstream_gene_variantCTtranslocase of inner mitochondrial
membrane 23 homolog B
0.0286
chr10129179426ENSG00000150760DOCK110q26.2rs7099958intron_variantTCdedicator of cytokinesis 10.0286
chr113078536ENSG00000110619CARS11p15.4rs4758463intron_variantCGcysteinyl-tRNA synthetase0.0286
chr12122079189ENSG00000182500ORAI112q24.31rs3741595synonymous_variantCTORAI calcium release-activated calcium
modulator 1
0.0286
chr12131623850ENSG00000111452ADGRD112q24.33rs35160436non-synonymous SNVAACadhesion G protein-coupled receptor D10.0286
chr13113793849ENSG00000126218F1013q34rs3211770upstream_gene_variantGAcoagulation factor X0.0286
chr1435228090ENSG00000198604BAZ1A14q13.1rs61981202intron_variantGAbromodomain adjacent to zinc finger
domain 1A
0.0286
chr1435237874ENSG00000198604BAZ1A14q13.1rs61981228downstream_gene_variantCAbromodomain adjacent to zinc finger
domain 1A
0.0286
chr1435483882ENSG00000100883SRP5414q13.2rs13379372sequence_featureACsignal recognition particle 54kDa0.0286
chr1435492299ENSG00000100883SRP5414q13.2rs4982254upstream_gene_variantAGAsignal recognition particle 54kDa0.0286
chr1435492301ENSG00000100883SRP5414q13.2rs80306194upstream_gene_variantCTTGTTATT
AGTTAACAG
Csignal recognition particle 54kDa0.0286
chr1435497285ENSG00000100883SRP5414q13.2rs78609489intron_variantTCsignal recognition particle 54kDa0.0286
chr162906934ENSG00000263325LA16c-325D7.116p13.3rs732532upstream_gene_variantGAprotease, serine 220.0286
chr163021417ENSG00000127564PKMYT116p13.3rs79505645upstream_gene_variantGTprogestin and adipoQ receptor family
member IV
0.0286
chr1615126890ENSG00000179889PDXDC116p13.11rs12926897upstream_gene_variantCTpyridoxal-dependent decarboxylase
domain containing 1
0.0286
chr1615850204ENSG00000133392MYH1116p13.11rs2272554synonymous_variantAGmyosin, heavy chain 11, smooth muscle0.0286
chr1615853596ENSG00000133392MYH1116p13.11rs2280764intron_variantCGmyosin, heavy chain 11, smooth muscle0.0286
chr1616138322ENSG00000103222ABCC116p13.11rs246221synonymous_variantTCATP binding cassette subfamily C member 10.0286
chr1616139714ENSG00000103222ABCC116p13.11rs35587synonymous_variantTCATP binding cassette subfamily C member 10.0286
chr1616139878ENSG00000103222ABCC116p13.11rs35588splice_region_variant
and intron_variant
AGATP binding cassette subfamily C member 10.0286
chr1757951973ENSG00000108423TUBD117q23.1rs2250526synonymous_variantGAtubulin delta 10.0286
chr1757992145ENSG00000241913RP5-1073F15.117q23.1rs3066247downstream_gene_variantTATCTribosomal protein S6 kinase B10.0286
chr1758037374ENSG00000189050RNFT117q23.1rs12600680upstream_gene_variantTCring finger protein, transmembrane 10.0286
chr1758042126ENSG00000189050RNFT117q23.1rs76419616upstream_gene_variantTCTBC1D3P1-DHX40P1 readthrough,
transcribed pseudogene
0.0286
chr1933882222ENSG00000124299PEPD19q13.11rs17569synonymous_variantGApeptidase D0.0286
chr2223487533ENSG00000100228RAB3622q11.22rs14764415_prime_UTR_variantCTRAB36, member RAS oncogene family0.0286
Table 4. Gene Set Enrichment Analysis (GSEA) for ALPK2.
Table 4. Gene Set Enrichment Analysis (GSEA) for ALPK2.
GeneSetsNESNOM p-valFDR q-val
Reactome WNT Ligand Biogenesis and Trafficking1.410.1100.398
PID WNT Signaling Pathway1.400.1040.221
WNT Up.V1 Up1.150.2490.356
WNT Up.V1 DN0.900.5660.582
Reactome Beta Catenin Independent WNT Signaling−3.180.0000.000
Reactome Signaling By WNT−3.070.0000.000
WP WNT Signaling Pathway−2.070.0040.014
Hallmark WNT Beta Catenin Signaling−2.060.0040.012
PID WNT Canonical Pathway−1.970.0100.014
Biocarta WNT Pathway−1.740.0230.043
PID WNT Noncanonical Pathway−1.700.0250.045
KEGG WNT Signaling Pathway−1.540.0500.081
WP WNT Signaling−1.260.1850.235
WNT Signaling−1.210.2410.257
Reactome Signaling by WNT In Cancer−1.200.2420.241
GOCC Catenin Complex1.820.0120.066
GOMF WNT Protein Binding0.900.5860.736
GOBP Cell Cell Signaling By WNT−2.610.0000.000
GOBP Regulation of WNT Signaling Pathway−2.240.0000.003
HP Downturned Corners of Mouth−2.170.0000.005
GOBP Positive Regulation of WNT Signaling Pathway−2.060.0000.009
GOBP Canonical WNT Signaling Pathway−2.050.0000.010
GOBP Negative Regulation of Canonical WNT Signaling Pathway−1.940.0080.017
GOBP Negative Regulation of WNT Signaling Pathway−1.800.0110.032
GOBP Positive Regulation of Canonical WNT Signaling Pathway−1.770.0210.036
GOMF Beta Catenin Binding−1.550.0570.090
GOBP Non-canonical WNT Signaling Pathway−1.510.0630.105
GOBP Regulation of Non-canonical WNT Signaling Pathway−1.250.1950.253
GOMF WNT Receptor Activity−1.170.2370.315
GOBP Regulation of WNT Signaling Pathway Planner Cell Polarity Pathway0.960.4870.544
NES = Normalized enriched score; NOM p-val = Sta:s:cally significant pathways p < 0.05; FDR qval = FDR adjusted p-value < 0.05.
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

Wei, J.; Wu, B.J.; Daoud, S.S. Whole-Exome Sequencing (WES) Reveals Novel Sex-Specific Gene Variants in Non-Alcoholic Steatohepatitis (MASH). Genes 2024, 15, 357. https://doi.org/10.3390/genes15030357

AMA Style

Wei J, Wu BJ, Daoud SS. Whole-Exome Sequencing (WES) Reveals Novel Sex-Specific Gene Variants in Non-Alcoholic Steatohepatitis (MASH). Genes. 2024; 15(3):357. https://doi.org/10.3390/genes15030357

Chicago/Turabian Style

Wei, Jing, Boyang Jason Wu, and Sayed S. Daoud. 2024. "Whole-Exome Sequencing (WES) Reveals Novel Sex-Specific Gene Variants in Non-Alcoholic Steatohepatitis (MASH)" Genes 15, no. 3: 357. https://doi.org/10.3390/genes15030357

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