View Article

Abstract

The etiology and clinical manifestations of immune-mediated diseases are known to be attributed to a variety of hereditary and environmental variables. In the instance of the genetic factor, genetic variations have been identified in a range of autoimmune disorders. Genome-wide association studies (GWAS) are a viable approach for determining the inherited basis of disease-causing genes. We can investigate mutation or polymorphisms correlated to identify the diseases and predict outcomes by fully understanding these results. Autoimmune diseases are distinguished by a lack of self-tolerance, which results in immune-mediated tissue damage and chronic inflammation. Although studies indicate associations between single nucleotide polymorphism (SNPs) in the human genome and autoimmune disorders, the findings are still ambiguous. A literature review was explored to identify the relationship between polymorphisms and autoimmune disease. In this study, we will discuss the significant outcomes of GWAS in common autoimmune conditions such as multiple sclerosis, diabetes, migraine, Parkinson's disease, and Alzheimer's disease. We further explore the prospective role of these innovations in terms of disease forecasting, basic biology, and the development of possible therapies. Continuous developments in technology and analytic methods have boosted the effectiveness and efficiency of genetic mapping approaches.

Keywords

GWAS, NonSynonymous, PD, AD, CRISPR-Cas9, WGHS, IPDGC

Introduction

The human genome innovation is becoming more accessible owing to breakthroughs in DNA sequencing. Due to the massive variety of mutations, interpreting genetic variation is one of the main issues facing genomics research nowadays [1]. The simplest form of polymorphism is a single-nucleotide polymorphism (SNP), which is caused by a single base mutation that changes one nucleotide for another. The insertion or deletion of a DNA sequence may result in a variety of further alterations [2, 3]. The insertion or deletion of a DNA sequence may result in a variety of further alterations. The 99.5% genomic similarity shared by humans suggests that epigenetic mutations and the rest of the 0.5% difference account for phenotypic variability [4] are sometimes referred to as mutations and often as polymorphisms [3, 5]. Any change from the norm in a DNA sequence is referred to as a mutation [6]. Conversely, polymorphism is an alteration in the genetic sequence of the population. A particular genetic variant is depicted in Figure 1A as a polymorphic modification. The majority of SNPs are biallelic (two alternative bases occur), require a minimum frequency in the population (>1%), and multiallelic SNPs do exist[7]. The aforementioned SNPs may impact the genetic code that affects either the coding (exons), intergenic, or non-coding (introns) domains [8, 9]. Polymorphisms with transitions (A ? G or C ?T) are relatively common as SNPs with transversions (A ? C/T; and G ?C / T) [10]. The majority of the genetic differences in the human genome, which comprise between 3 and 5 million in each person and involve a single base change in the alleles, are known SNPs [11, 12]. The existence of short and variable tandem repeats, insertion or deletion polymorphisms (InDels), and SNPs causes sequence modifications. Many InDels occur in functionally significant regions of the DNA and may thus play a role in disease progression [13]. Human genes correlated with various multifaceted disorders were found using candidate genes or GWAS, highlighted in Table 1 [14]. Through the entire genome, mutations occur at a rate of one in a thousand (roughly every 300-2000) pair of bases, while the majority of significant SNPs occur in coding domains (about 50 thousand) [12]. The human genome has an estimated 84.7 million polymorphisms [15]. In one study, around 50% of variants occur in non-coding regions, 25% result in missense variants and the other are silent mutations, may not affect transcribed  protein [16].



       
            Picture1.png
       

    
       
            Picture2.png
       

    

Figure 1. A) A diagram depicting a single nucleotide polymorphism (SNP) B) The specific categorization of variations


There are primarily two types of SNPs found among them as follows: synonymous SNPs (sSNPs) and non-synonymous (nsSNP). The precise categorization of variants is illustrated in Figure 1B [2]. As such, a large number of variations from the human genome project (HGP) are currently accessible. In the last 5 years, over 500 GWASs have revealed thousands of DNA variants on more than 300 diseases and traits that are functionally characterized [17]. Human genes correlated with various multifaceted disorders were found using candidate genes or GWAS, highlighted in Figure 2 [14]. Prior studies showed that nsSNPs contribute to about half of the alterations correlated with many genetic disorders, including inflammatory and autoimmune disorders [17].


Table 1. A set of genes attributed to the probability of developing various multifaceted human disorders.


       
            Screenshot 2024-09-08 130234.png
       

    


The present study showed that nsSNPs, notably missense SNPs have gained attention as potential variables in disease association research with complicated health issues and phenotypic characteristics. A SNP innovation is often used to detect genes associated with diseases and examine inter-individual differences in therapy responsiveness [16]. The conclusion of the entire genome project has led to renewed interest in biological sciences. It offers the techniques for understanding the biological basis of genetic variations, the most frequent inherent trait, evolution, intricate and common diseases such as overweight, hypertension, diabetes, and schizophrenia, as well as to produce gene-based therapeutic medicines [18]. The modification in genomic sequences is thought to be a factor in the evolution of the genome. It may be possible to improve genome-based research on human risk for certain diseases, generate more effective and safer food and drugs for patients, and comprehend how evolution works by categorizing genetic variants across various groups and species. Even so, researchers acknowledge that prior SNP innovation will have an effect on medicine; it must conquer a variety of limitations [16, 19].


       
            Picture3.png
       

    Figure 2. Functional representation of SNPs.


Functional categorization of polymorphisms

Functional variants are categorized based on the region where they are mapped and Table 2 shows regulatory SNPs (rSNPs) and microRNA regulatory SNPs (miR-rSNPs), type of functional SNPs located in coding and non-coding genes, respectively; both variants alter gene regulation [20, 21]. The functional changes found in parent mRNA form (pre-mRNA) and mature mRNA are referred to as structural RNA SNPs (srSNPs), whilst microRNA srSNPs are termed as miR-srSNPs. Although srSNPs regulate mRNA translation, splicing, structure and stability, protein maturation, function, and miRNA-mRNA interaction [20, 22]. The primary miRNA and pre-miRNA transcript processing, splicing, miRNA, and mRNA binding and function are all affected by miR-srSNPs [23]. In contrast, nsSNPs are classified to be nonsense or missense as illustrated in Figure 2; the former results in a stop codon and a premature end to the amino acids, whereas the latter results in a protein alteration. Both possess a significant impact, but the latter may not be as severe if substitute amino acids have identical chemical compositions and biological abilities. Both forms of variants have an impact on protein sequence, configuration, and activity [14].


Table 2. Functional characterization of SNPs

        
            Screenshot 2024-09-08 130333.png
       

    


Significance of SNPs in Genome Detection

Many complex traits exhibit quantitative variation, and the locus controlling these traits is known as a quantitative trait locus (QTL) [24]. By identifying the entire DNA loci that cause sophisticated variations in traits, lacking a biological map built with genetic markers. With the advent of different identification of sequence variation methods and genotyping, studies utilizing SNPs have become more effective. Polymorphism recognition can be divided into screening genetic sequences for unknown mutations, and assessing (genotyping) people for identified variants. The pursuit of novel mutations can be defined into two approaches: global (random) and localized (targeted) methods [25]. Furthermore, before sequence determination, SNPs must be monitored; several approaches for prescreening SNPs, including single-strand structural variation, denatured dynamics, chemical cleavage, enzyme splitting, array hybridization, repair of mismatches, and bacteriophage Mu DNA transposition are just a few examples [16, 26-30]. The fundamental disadvantage of these approaches is the demand for prior sequence knowledge, which implies the region for the design of primers should be simple. Even so, these approaches are bound to extremely similar areas where the sequence is obtained through homologous cloning. Methods for exploiting variants at random across the genome, such as representation shotgun sequencing, primer ligation-mediated PCR, and degenerate oligonucleotide-primed PCR, were formed [31, 32]. Despite the benefit of having fewer duplicates, such approaches still require sequencing millions of clones to provide evidence suitable for mutation research, and efficacy was substantially lower than anticipated. As such, there remains significant space for development in SNP identification tools, precision, sensibility, and effectiveness.

Autoimmune disease-associated genes

Immune-related diseases, including celiac disease, inflammatory bowel disease, heart disease, arthritis, diabetes, and multiple sclerosis, are a variety of biologically complex disorders that result from autoimmune malfunction and share basic pathogenic processes that remain unclear, although it is commonly believed to develop as a result of an imbalance interaction of environmental as well as genetic variables [33, 34]. It exhibits abnormalities of several significant regulatory processes, and methods such as GWAS along with NGS have greatly enhanced our awareness of biological factors [35]. Over decades, GWAS studies identified a numbers of susceptible alleles, both prevalent and pathogen-specific. Also, Immunochip platform analyzed 200,000 genetic variations in 186 autoimmune related disorder, revealing typical vulnerable loci for several of the aforementioned diseases [36, 37].  Autoimmune disorders arise when the body defense system is misdirected towards the host [38], they vary from organ-specific disorders in which antibodies and T cells react to self-antigens [39]. In regard to the process of delayed immune reactions in the onset of autoimmune disorders, aberrant immune actions are involved in the progression of the diseases. Clinical studies showed that autoimmunity disorders affects around 5-8% of the world's population with limited therapies. Over 80 immune-related diseases were found, and they are seen as a serious global socioeconomic issue since they cause patients severe suffering and reduce their standard of existence [38, 39]. Despite the etiology of many such diseases being unknown, the discovery of complex aspects involving pathogenetic, inherited, and ecological regulators could contribute to the emergence of novel therapies, prior screening ability, and the perception of autoimmune processes that underlie these disorders [40, 41]. New research shows that genomic data may assist in comprehending the etiology of immune-mediated diseases and give a genetic predisposition by discovering the genes and particular processes responsible for the disease. The HLA genes, for example, were identified as the most potent and early-risk genes associated with autoimmunity and predict the extent of the disorder that responds to biological drugs [42]. In this review, we will highlight the noteworthy outcomes of GWAS in common autoimmune diseases as discussed below. We explore the potential relevance of these breakthroughs in disease forecasting, and basic biology, and explore how many currently known genetic mutations will probably be revealed to influence disease vulnerability.

Type 2 Diabetes Mellitus (T2DM)

Diabetes is a significant chronic condition that endangers human life. In 2017, there were an estimated 451 million diabetes sufferers globally [43]. Based on the International Diabetes Federation (IDF) research, around 415 million people have diabetes, with T2D contributing to 90% of these cases [44]. Globally, diabetes was correlated with CDKAL1, PRKAA2, ABCA1, FADS, HLA-B, TCF7L2, IGF2BP2, and EXT2 genes. Currently, preferable 20 SNPs related to SLC30A8 (rs13266634), CDKN2A/2B (rs10811661), HHEX (rs1111875), and TCF7L2 (rs7903146) serving major roles in the risk of T2DM in the European population [45-47]. The Human Leukocyte Antigen class B (HLA-B) gene was related to T2D in the study cohort. The data reveal a substantial link between SNP rs2308655 and T2D in the Pashtun tribal population [48]. The other HLA alleles, mainly rs1051488, rs1131500, rs1050341, and rs1131285, were identified. A study issued in biology discovered rs560887 in the G6PC2 gene was linked with FBG while not with T2DM risk, and multiple studies stated that variations in the G6PC2 gene were linked to T2DM susceptibility. Another study found that G6PC2 (rs16856187) and GCKR haplotypes were related to T2DM vulnerability [49], and also revealed rs492594, and rs2232328 variants in Arabic people associated with diabetes [50, 51]. The SNPs of glucokinase (GCK) rs1799831 were linked with gestational diabetes mellitus (GDM) in the Indian population. A T2DM was related to rs1276891 and 44,184,184 3?UTR in the GCK gene in the Indian population [52]. Also, GCKR rs780094 and rs1260326 alleles have been linked to a lower risk of T2DM and obesity in the Han Chinese group. Fasting hyperglycemia was found to be molded by a gene/gene interaction between rs780094 and rs1799884 [53]. According to Sadeghi et al. (2021) found a correlation between rs28514894 and rs2303044 in the NR1H2 gene and a chance of developing T2D [54]. Mutations in the 3'UTR of SLC30A8 (rs2466293 and rs2466294) were shown to increase Iranian susceptibility to T2D by influencing the binding location of certain miRNAs and decreasing the stability of SLC30A8 mRNA transcripts [55]. Early detection of this genetic marker would allow health professionals to provide adequate guidance and therapy to patients, allowing for frequent follow-up and modification of lifestyle risk factors and daily meal intakes to protect them from developing T2DM at an early stage [56].

Multiple sclerosis (MS)

Multiple sclerosis is a prolonged, degenerative disorder of the central nervous system characterized by inflammatory and persistent neuronal degeneration. The HLA class II, ApoE, IL-1ra, IL-1beta, TNF-alpha, TNF-beta, and CCR5 genes have received tremendous research interest to explore disease-modifying implications in MS [57]. Currently, two polymorphisms found in the IL2RA gene, rs2104286 and rs12722489, SOCS-1 gene (rs243324), IL-16 (rs4072111 C/T), TNF-alpha-308 G/A (rs1800629 G/A), and IL-18-607 C/A (rs1946518 C/A) and rs352162 and rs187084 markers is regarded as a potential risk of MS [58]. The SIRT1 gene was studied in patients with vision impairment and MS that correlates with rs3818292, rs3758391, and rs7895833 substitutions. It also has a part in repairing DNA, mitochondrial formation, and cellular death. The potential contrast of CD58 variants, particularly rs2300747, rs12044852, and rs1335532, with MS among the Malay inhabitants of Malaysia was studied [59]. An analysis of six mutations comprising the MIR137HG (rs1625579), GAS5 (rs2067079), MIR3142HG (rs57095329), MIR146A (rs2910164), MIR155HG (rs767649), and IRAK1 (rs3027898) genes found an association with MS. A new study linked the variant rs12959006 found in the myelin basic protein (MBP) gene to a greater probability of relapsing and an adverse outcome [60].

Migraine

Migraines are a common neurovascular disorder with a complex of between 40-57% hereditary etiology, migraine without aura (MO) and migraine with aura (MA) are the two most common subtypes [61]. Polymorphisms in the ACE, DBH, TRPM8, COMT, GABRQ, CALCA, TRPV1, and other genes were identified as affecting migraine susceptibility.[62] Modern GWAS research identified 4 variants on chromosomal location 8q22.1, 2q37.4, 12q13.3, and 1p36.32 that have a strong association with migraine subtypes [63]. The most recent GWAS for migraine found 38 loci, and to determine whether 46 SNPs (most common rs12135062, rs10166942, rs11031122, rs11172113, and rs17857135) at these loci have a strong association with migraine. According to the Women's Genome Health Study (WGHS), rs11031122 variations were found as variables by examining its link with MA and MO in meta-analyses [64, 65]. The first meta-analysis conducted by Anttila et al. included 29 research studies of 23,285 migraine sufferers and 95,425 controls [66]. The glutamatergic nerve signaling loci were (rs1835740) MTDH, (rs11172113) LRP1, and (rs3790455) MEF2D. For a synaptic growth and neural plasticity genes include (rs6478241) ASTN2, (rs1320832) FHL5, and sensitive to pain involve (rs10166942) TRPM8 gene and for metallic proteins include (rs10504861) MMP16, (rs10915437) AJAP1, (rs12134493) TSPAN2 genes, 5) and for the circulation and metabolites have (rs4379368) C7orf10, (rs2651899) PRDM16, (rs9349379) PHACTR1, and (rs7640543) TGFBR2 genes [67-69]. The rs1835740 SNP is found in the 8q22.1 chromosome, in the MTDH and PGCP gene related to glutamate metabolism and found that raising the release of glutamate or lower uptake raises the probability of migraine attacks.[70] The MTHFR genetic variants rs1801133 (C66T) and rs1801131 (A1298C) are risk factors for migraine susceptibility and are primarily linked to high homocysteine concentrations. The rs7590387, rs3754701 substitutions around the RAMP1 gene associated with migraine risk in Asian and Caucasian individuals were assessed [71].

Parkinson's disease (PD)

Several researchers have found that multiple genes affect PD vulnerability and the pathology remains believed to be solely environmental. Several polymorphisms were investigated by Australian Parkinson's Disease Registration (APDR) such as CD14 (rs2569190), MUC1 (rs4072037), MUC2 (rs11825977), CLDN2 (rs12008279 and rs12014762), and CLDN4 (rs8629). The PGLYRP4 (rs10888557) genotype significantly increased the risk of PD in each group. The PGLYRP2 (rs892145), TLR1 (rs4833095) or TLR2 (rs3804099), and MUC2 (rs11825977) variations substantially increased PD disease susceptibility in the APDR cohort. Five unknown loci (ACMSD, STK39, MCCC1/LAMP3, SYT11, and CCDC62/HIP1R) were found, and six were already known loci (MAPT, SNCA, HLADRB5, BST1, GAK, and LRRK2) were identified [72-77]. Another 5 PD-associated loci such as PARK16, STX1B, FGF20, STBD1, and GPNMB were identified by the IPDGC [63]. Most detailed meta-analyses included results from 7 million SNPs from GWAS and smaller-scale PD association studies. Future research must focus on identifying functional variations and comprehending the molecular implications of each risk locus.

Alzheimer’s disease

By 2050, there will probably be 152 million cases of dementia worldwide, with AD being the most prevalent form of autoimmune disorder that is regarded as the more frequent basis for cognitive impairment and contributes to 70% of AD instances [78]. The amino acid at position 69 shifts from arginine to glycine (R69G) as a result of the minor allele at rs2455069. The CD33 SNP rs2455069 is strongly linked with dementia in Italy and the R69G amino acid shift modifies the structure of CD33 and affects its susceptibility to sialic acid residue [79]. Moreover, it was found that the ABCA7 variants rs3764650 and rs4147929 were associated with AD cases. Up to 20 risk genes were identified and described by prior GWAS investigations, including ABCA7, BIN1, CLU, CR1, PICALM, SORL1, and others [80]. New studies have revealed that the mutations in GRN, TMEM106B, Complement C7, and RBFOX1 are linked to AD in different populations [81]. The rs3810950 polymorphism may have a small but significant effect on the risk of AD in the Czech people, demonstrated by the link between the genotype of choline acetyltransferase (ChAT) and a 1.25 times greater risk of AD [82]. Furthermore, there was considerable discussion regarding the correlation between the APOC1 rs11568822 SNP and the probability of AD in many studies [83].

FUTURE PERSPECTIVE & CHALLENGES

The research described above gives a summary of significant mutations related to autoimmune and multifaceted genetic disorders. Despite significant collaborative efforts, many disorders can no longer be studied inherently, and the heritability of complex diseases is only partially explained by GWAS studies. The risk gene prognosis may be improved by more research, including novel approaches for finding rare modifications using DNA sequencing and structural variants. To efficiently exploit the GWAS tool to improve the knowledge about cancer physiology and discover potential targets for clinical and therapeutic methods, it is necessary to ascertain the functional data implications. Concerning neurodegeneration, heart disease, or immunological diseases, these results may indicate a variety of provided inherited and mechanistic variables. Many researchers are exploring new filtering techniques, effective computational methods, and process methods for studying SNP interactions. Although metagenome as collecting data for massive cohorts has risen greatly, it is still difficult to analyze the microbiome about host DNA. The effects of exogenous and external variables on the gut microbiome, which may obscure the effects of genetic variations; the intricate organization of microbiome data, makes analysis tough; the substantial number of tests done are challenging, which necessitates the collection of large cohorts to address the issue of various assessment. In future decades, we believe that introducing the findings of GWAS will increase the probability of effective biological and therapeutic discoveries in neurology. A high-dimensional difficulty, computing constraints, the absence of marginal effects, lacking heritability, and genetic variability are among the limitations.


       
            Picture4.png
       

    Figure 4. Taxonomy of mutations according to a sequenced concept in the pairing and non-paired approach [84].


Several machine learning and data mining techniques were used to address such issues. Research and development of economic or breeding features related to these genes, as well as the invention of genetic breeding methods using DNA loci, are all significant to genetic engineering. In the era of Next Gen Sequencing (NGS) and precise medicine, the whole genome or transcriptome sequencing methods are projected to grow considerably less laborious and cost-effective. Multiple global collaborations, such as ENCODE (Encyclopaedia of DNA elements) and HapMap (Haplotype Map), have followed to map the DNA variations and regulatory elements of a human genome [84]. To distinguish between the mutation and polymorphism of genomic variation, we propose an obvious difference between an SNP and a somatic mutation, as shown in Figure 3. It is challenging to establish effective method-based strategies for predicting the risk of infectious diseases. Researchers can use powerful computational methods to analyze high-speed sequencing programs. Thus far, new genetic technology allowed the merging of molecular genetics data with qualitative genetic research formerly available in the discipline. Some processes, such as methylation of cytosine and histone modification, have now become innovations that help to comprehend how certain active SNPs function in certain ambient conditions. Another approach such as DNA editing on designed nucleases (Crispr-Cas9, Crispr/ScCas9, and Crispr-Cas12) [85-87], may be used later on to modify genes using new targeted methods. The significance of gene mapping strategies has risen as technology and analytic tools have advanced. Such investigations complement genetic methods and are comparable to human genome sequencing because they allow the production of a vast place of 'functional' facts that can be systematically explored to find genes encoding proteins that will likely have a key part in the progression of diseases.

CONCLUSION

Genetic mapping of mutant genes in diseases with more complex genetic aetiologies, as well as modifier genes, has become more possible. In the future, many genetic variants that are currently known will probably be found to affect the extent of immune-mediated disorders in both prevalent and rare circumstances. Such understanding will allow for more accurate disease outcome prediction in individuals, as well as more targeted preventive measures. A systematic analysis of known genetic variants with no currently defined functional effects will almost certainly lead to the discovery of a subgroup that influences drug response. It is feasible to pinpoint genetic characteristics that increase the chance of negative responses, as well as factors affecting the effectiveness and proper medication. These studies reinforce biological methods and are comparable to genome sequencing as they can enable the production of a massive store of functional data that can be systematically mined to find genes encoding proteins that anticipate playing a critical role in disease progression.

ACKNOWLEDGMENTS

We thank all the authors who have contributed to this review paper.

AUTHOR CONTRIBUTION

All authors discussed the results and contributed to the final manuscript.

FUNDING STATEMENT

No specific funding for this research was received from any public, commercial, or not-for-profit sectors.

CONFLICT OF INTEREST

No conflict of interest

PARTICIPANT CONSENT AND ETHICAL APPROVAL

 Not applicable.

RIGHTS FOR PUBLICATION

 The work has been authorized for submission by all authors.

COMPETING INTERESTS

The authors disclose that they have no conflicts of interest.

REFERENCES

  1. Vallejos-Vidal, E., et al., Single-nucleotide polymorphisms (SNP) mining and their effect on the tridimensional protein structure prediction in a set of immunity-related expressed sequence tags (EST) in Atlantic salmon (Salmo salar). Frontiers in genetics, 2020. 10: p. 1406.
  2. Ali, M.Z., et al., In Silico Analysis Identified Putative Pathogenic Missense nsSNPs in Human SLITRK1 Gene. Genes, 2022. 13(4): p. 672.
  3. Fucharoen, S., Genetic polymorphisms and implications for human diseases. J Med Assoc Thai, 2007. 90(2): p. 394-8.
  4. Yudianto, A., et al., Allelic sharing among madurese as a tool of madurese identification using 11 short tandem repeats and amelogenin gene: An observational analytical study. Journal of International Oral Health, 2021. 13(1): p. 89.
  5. Shastry, B.S., SNPs: impact on gene function and phenotype. Single nucleotide polymorphisms: Methods and protocols, 2009: p. 3-22.
  6. Wei, T., et al., A novel multiplex assay of SNP-STR markers for forensic purpose. PLoS One, 2018. 13(7): p. e0200700.
  7. Wang, Z., et al., Systematic identification and analysis of exonic splicing silencers. Cell, 2004. 119(6): p. 831-845.
  8. Ahmad, T., M.A. Valentovic, and G.O. Rankin, Effects of cytochrome P450 single nucleotide polymorphisms on methadone metabolism and pharmacodynamics. Biochemical pharmacology, 2018. 153: p. 196-204.
  9. Van Dijk, E.L., et al., Ten years of next-generation sequencing technology. Trends in genetics, 2014. 30(9): p. 418-426.
  10. Edwards, D., et al., What are SNPs? Association mapping in plants, 2007: p. 41-52.
  11. Dakal, T.C., et al., Predicting the functional consequences of non-synonymous single nucleotide polymorphisms in IL8 gene. Scientific reports, 2017. 7(1): p. 6525.
  12. Collins, F.S., L.D. Brooks, and A. Chakravarti, A DNA polymorphism discovery resource for research on human genetic variation. Genome research, 1998. 8(12): p. 1229-1231.
  13. Mullaney, J.M., et al., Small insertions and deletions (INDELs) in human genomes. Human molecular genetics, 2010. 19(R2): p. R131-R136.
  14. Ramírez-Bello, J. and M. Jiménez-Morales, Functional implications of single nucleotide polymorphisms (SNPs) in protein-coding and non-coding RNA genes in multifactorial diseases. Gaceta medica de Mexico, 2017. 153(2): p. 238-250.
  15. Chauhan, W., et al., Cataloging the potential SNPs (single nucleotide polymorphisms) associated with quantitative traits, viz. BMI (body mass index), IQ (intelligence quotient) and BP (blood pressure): an updated review. Egyptian Journal of Medical Human Genetics, 2022. 23(1): p. 57.
  16. Shastry, B.S., SNPs in disease gene mapping, medicinal drug development and evolution. Journal of human genetics, 2007. 52: p. 871-880.
  17. Chen, R., et al., Non-synonymous and synonymous coding SNPs show similar likelihood and effect size of human disease association. PloS one, 2010. 5(10): p. e13574.
  18. Emilien, G., et al., Impact of genomics on drug discovery and clinical medicine. Qjm, 2000. 93(7): p. 391-423.
  19. Gonzaga-Jauregui, C., J.R. Lupski, and R.A. Gibbs, Human genome sequencing in health and disease. Annual review of medicine, 2012. 63: p. 35-61.
  20. Ramírez-Bello, J., G. Vargas-Alarcón, and J.M. Fragoso, Single nucleotide polymorphisms (SNPs): functional implications of regulatory-SNP (rSNP) and structural RNA (srSNPs) in complex diseases. Gaceta medica de Mexico, 2013. 149(2): p. 220-228.
  21. Sobieszczyk, M.E., J.R. Lingappa, and M.J. McElrath, Host genetic polymorphisms associated with innate immune factors and HIV-1. Current Opinion in HIV and AIDS, 2011. 6(5): p. 427-434.
  22. Sadee, W., et al., Pharmacogenomics of the RNA world: structural RNA polymorphisms in drug therapy. Clinical Pharmacology & Therapeutics, 2011. 89(3): p. 355-365.
  23. Sun, G., et al., SNPs in human miRNA genes affect biogenesis and function. Rna, 2009. 15(9): p. 1640-1651.
  24. Albert, F.W. and L. Kruglyak, The role of regulatory variation in complex traits and disease. Nature Reviews Genetics, 2015. 16(4): p. 197-212.
  25. Kwok, P.-Y. and X. Chen, Detection of single nucleotide polymorphisms. Current issues in molecular biology, 2003. 5(2): p. 43-60.
  26. Yue, P. and J. Moult, Identification and analysis of deleterious human SNPs. Journal of molecular biology, 2006. 356(5): p. 1263-1274.
  27. Oleykowski, C.A., et al., Mutation detection using a novel plant endonuclease. Nucleic acids research, 1998. 26(20): p. 4597-4602.
  28. Goldrick, M.M., RNase cleavage?based methods for mutation/SNP detection, past and present. Human Mutation, 2001. 18(3): p. 190-204.
  29. Sokurenko, E.V., et al., Detection of simple mutations and polymorphisms in large genomic regions. Nucleic Acids Research, 2001. 29(22): p. e111-e111.
  30. Orsini, L., et al., SNP discovery by mismatch-targeting of Mu transposition. Nucleic Acids Research, 2007. 35(6): p. e44.
  31. Janiak, A., et al., Application of degenerate oligonucleotide primed PCR (DOP-PCR) for SNP discovery in soybean. Euphytica, 2008. 162: p. 249-256.
  32. Xu, J.-Y., G.-B. Xu, and S.-L. Chen, A new method for SNP discovery. BioTechniques, 2009. 46(3): p. 201-208.
  33. Cotsapas, C. and D.A. Hafler, Immune-mediated disease genetics: the shared basis of pathogenesis. Trends in immunology, 2013. 34(1): p. 22-26.
  34. Wu, X., H. Chen, and H. Xu, The genomic landscape of human immune-mediated diseases. Journal of human genetics, 2015. 60(11): p. 675-681.
  35. Diaz-Gallo, L.-M. and J. Martin, Common genes in autoimmune diseases: a link between immune-mediated diseases. Expert Review of Clinical Immunology, 2012. 8(2): p. 107-109.
  36. Parkes, M., et al., Genetic insights into common pathways and complex relationships among immune-mediated diseases. Nature Reviews Genetics, 2013. 14(9): p. 661-673.
  37. Baune, B.T., et al., Interleukin-6 gene (IL-6): a possible role in brain morphology in the healthy adult brain. Journal of neuroinflammation, 2012. 9(1): p. 1-9.
  38. Fugger, L., L.T. Jensen, and J. Rossjohn, Challenges, progress, and prospects of developing therapies to treat autoimmune diseases. Cell, 2020. 181(1): p. 63-80.
  39. Wang, L., F.S. Wang, and M.E. Gershwin, Human autoimmune diseases: a comprehensive update. Journal of internal medicine, 2015. 278(4): p. 369-395.
  40. Selmi, C., Autoimmunity in 2019. Clinical Reviews in Allergy & Immunology, 2020. 59(3): p. 275-286.
  41. Chen, J.-Q., P. Szodoray, and M. Zeher, Toll-like receptor pathways in autoimmune diseases. Clinical reviews in allergy & immunology, 2016. 50: p. 1-17.
  42. Zeggini, E., et al., Translational genomics and precision medicine: Moving from the lab to the clinic. Science, 2019. 365(6460): p. 1409-1413.
  43. Cho, N.H., et al., IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes research and clinical practice, 2018. 138: p. 271-281.
  44. Atlas, D., IDF diabetes atlas. International Diabetes Federation (9th editio). Retrieved from http://www. idf. org/about-diabetes/facts-figures, 2019.
  45. Sladek, R., et al., A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature, 2007. 445(7130): p. 881-885.
  46. Scott, L.J., et al., A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. science, 2007. 316(5829): p. 1341-1345.
  47. Vatankhah Yazdi, K., et al., SLC30A8, CDKAL1, TCF7L2, KCNQ1 and IGF2BP2 are associated with type 2 diabetes mellitus in Iranian patients. Diabetes, Metabolic Syndrome and Obesity, 2020: p. 897-906.
  48. Jan, A., et al., Association of HLA-B Gene Polymorphisms with Type 2 Diabetes in Pashtun Ethnic Population of Khyber Pakhtunkhwa, Pakistan. Journal of Diabetes Research, 2021. 2021.
  49. Shi, Y., et al., Meta-analyses of the association of G6PC2 allele variants with elevated fasting glucose and type 2 diabetes. PLoS One, 2017. 12(7): p. e0181232.
  50. Hu, C., et al., A genetic variant of G6PC2 is associated with type 2 diabetes and fasting plasma glucose level in the Chinese population. Diabetologia, 2009. 52: p. 451-456.
  51. Li, C., et al., Glucose metabolism-related gene polymorphisms as the risk predictors of type 2 diabetes. Diabetology & Metabolic Syndrome, 2020. 12: p. 1-6.
  52. Muller, Y.L., et al., Common genetic variation in the glucokinase gene (GCK) is associated with type 2 diabetes and rates of carbohydrate oxidation and energy expenditure. Diabetologia, 2014. 57: p. 1382-1390.
  53. Qi, Q., et al., Association of GCKR rs780094, alone or in combination with GCK rs1799884, with type 2 diabetes and related traits in a Han Chinese population. Diabetologia, 2009. 52: p. 834-843.
  54. Sadeghi, M.B., et al., Significant association of LXR? (NR1H2) polymorphisms (rs28514894, rs2303044) with type 2 diabetes mellitus and laboratory characteristics. Journal of Diabetes & Metabolic Disorders, 2021. 20: p. 261-270.
  55. Sargazi, S., et al., SNPs in the 3?-untranslated region of SLC30A8 confer risk of type 2 diabetes mellitus in a south-east Iranian population: Evidences from case-control and bioinformatics studies. Journal of Diabetes & Metabolic Disorders, 2020. 19: p. 979-988.
  56. Ayelign, B., et al., TNF-? (? 308) Gene Polymorphism and Type 2 Diabetes Mellitus in Ethiopian Diabetes Patients. Diabetes, metabolic syndrome and obesity: targets and therapy, 2019: p. 2453-2459.
  57. Kallaur, A.P., et al., Genetic polymorphisms associated with the development and clinical course of multiple sclerosis. International journal of molecular medicine, 2011. 28(4): p. 467-479.
  58. Dominguez?Mozo, M., et al., Toll?like receptor?9 in S panish multiple sclerosis patients: an association with the gender. European Journal of Neurology, 2014. 21(3): p. 537-540.
  59. Ching, Y.M., et al., Association of CD58 polymorphism and multiple sclerosis in Malaysia: a pilot study. Autoimmunity Highlights, 2019. 10: p. 1-5.
  60. Hamid, M.M.A.E., et al., Multiple sclerosis: an associated single-nucleotide polymorphism study on Egyptian population. Network Modeling Analysis in Health Informatics and Bioinformatics, 2020. 9: p. 1-7.
  61. Esserlind, A.L., et al., Replication and meta?analysis of common variants identifies a genome?wide significant locus in migraine. European journal of neurology, 2013. 20(5): p. 765-772.
  62. Roshandel, D., et al., Genetic predisposition to ocular surface disorders and opportunities for gene-based therapies. The Ocular Surface, 2023.
  63. Tan, M.-S., et al., Genome-wide association studies in neurology. Annals of translational medicine, 2014. 2(12).
  64. Gormley, P., et al., Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nature genetics, 2016. 48(8): p. 856-866.
  65. Kossowsky, J., et al., Association of genetic variants with migraine subclassified by clinical symptoms in adult females. Frontiers in Neurology, 2021. 11: p. 617472.
  66. Lin, Q.-F., et al., Association of genetic loci for migraine susceptibility in the she people of China. The journal of headache and pain, 2015. 16(1): p. 1-6.
  67. Kaur, S., et al., Role of single nucleotide polymorphisms (SNPs) in common migraine. The Egyptian Journal of Neurology, Psychiatry and Neurosurgery, 2019. 55(1): p. 1-7.
  68. Genome-wide association study of migraine implicates a common susceptibility variant on 8q22. 1. Nature genetics, 2010. 42(10): p. 869-873.
  69. Klimov, E., et al., Genetics of migraine-is there any progress. J Neurol Stroke, 2017. 7(4): p. 00245.
  70. van den Maagdenberg, A.M., et al., Migraine: gene mutations and functional consequences. Current opinion in neurology, 2007. 20(3): p. 299-305.
  71. Kumar, Y.S., et al., Association of RAMP-1 gene polymorphisms and risk of migraine: Systemic review and meta-analysis. Human Gene, 2023: p. 201157.
  72. Gorecki, A.M., et al., Single nucleotide polymorphisms associated with gut homeostasis influence risk and age-at-onset of Parkinson's disease. Frontiers in aging neuroscience, 2020. 12: p. 603849.
  73. Goldman, S.M., et al., Peptidoglycan recognition protein genes and risk of Parkinson's disease. Movement Disorders, 2014. 29(9): p. 1171-1180.
  74. Marras, C., C.G. Canning, and S.M. Goldman, Environment, lifestyle, and Parkinson's disease: implications for prevention in the next decade. Movement Disorders, 2019. 34(6): p. 801-811.
  75. Burgueño, J.F. and M.T. Abreu, Epithelial Toll-like receptors and their role in gut homeostasis and disease. Nature Reviews Gastroenterology & Hepatology, 2020. 17(5): p. 263-278.
  76. de Matos Lourenço, C., et al., Characterization and strong risk association of TLR2 del-196 to-174 polymorphism and Helicobacter pylori and their influence on mRNA expression in gastric cancer. World Journal of Gastrointestinal Oncology, 2020. 12(5): p. 535.
  77. Theunissen, F., et al., Structural variants may be a source of missing heritability in sALS. Frontiers in Neuroscience, 2020. 14: p. 47.
  78. Mann, D.M., Pyramidal nerve cell loss in Alzheimer's disease. Neurodegeneration, 1996. 5(4): p. 423-427.
  79. Tortora, F., et al., CD33 rs2455069 SNP: correlation with alzheimer’s disease and hypothesis of functional role. International Journal of Molecular Sciences, 2022. 23(7): p. 3629.
  80. Kunkle, B.W., et al., Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates A?, tau, immunity and lipid processing. Nature genetics, 2019. 51(3): p. 414-430.
  81. Wang, L., et al., Analysis of Genetic Association Between ABCA7 Polymorphism and Alzheimer’s Disease Risk in the Southern Chinese Population. Frontiers in Aging Neuroscience, 2022. 14.
  82. Halova, A., et al., CHAT gene polymorphism rs3810950 is associated with the risk of Alzheimer’s disease in the Czech population. Journal of biomedical science, 2018. 25: p. 1-9.
  83. Zhou, Q., et al., Association between APOC1 polymorphism and Alzheimer’s disease: a case-control study and meta-analysis. PloS one, 2014. 9(1): p. e87017.
  84. Karki, R., et al., Defining “mutation” and “polymorphism” in the era of personal genomics. BMC medical genomics, 2015. 8: p. 1-7.
  85. Zhang, H., et al., Meta-analysis of GWAS on both Chinese and European populations identifies GPR173 as a novel X chromosome susceptibility gene for SLE. Arthritis research & therapy, 2018. 20(1): p. 1-8.
  86. Zhang, Y., et al., Applications and potential of genome editing in crop improvement. Genome biology, 2018. 19: p. 1-11.

Gosavi, G., et al., Applications of CRISPR technology in studying plant-pathogen interactions: overview and perspective. Phytopathology Research, 2020. 2(1): p. 1-9.

Reference

  1. Vallejos-Vidal, E., et al., Single-nucleotide polymorphisms (SNP) mining and their effect on the tridimensional protein structure prediction in a set of immunity-related expressed sequence tags (EST) in Atlantic salmon (Salmo salar). Frontiers in genetics, 2020. 10: p. 1406.
  2. Ali, M.Z., et al., In Silico Analysis Identified Putative Pathogenic Missense nsSNPs in Human SLITRK1 Gene. Genes, 2022. 13(4): p. 672.
  3. Fucharoen, S., Genetic polymorphisms and implications for human diseases. J Med Assoc Thai, 2007. 90(2): p. 394-8.
  4. Yudianto, A., et al., Allelic sharing among madurese as a tool of madurese identification using 11 short tandem repeats and amelogenin gene: An observational analytical study. Journal of International Oral Health, 2021. 13(1): p. 89.
  5. Shastry, B.S., SNPs: impact on gene function and phenotype. Single nucleotide polymorphisms: Methods and protocols, 2009: p. 3-22.
  6. Wei, T., et al., A novel multiplex assay of SNP-STR markers for forensic purpose. PLoS One, 2018. 13(7): p. e0200700.
  7. Wang, Z., et al., Systematic identification and analysis of exonic splicing silencers. Cell, 2004. 119(6): p. 831-845.
  8. Ahmad, T., M.A. Valentovic, and G.O. Rankin, Effects of cytochrome P450 single nucleotide polymorphisms on methadone metabolism and pharmacodynamics. Biochemical pharmacology, 2018. 153: p. 196-204.
  9. Van Dijk, E.L., et al., Ten years of next-generation sequencing technology. Trends in genetics, 2014. 30(9): p. 418-426.
  10. Edwards, D., et al., What are SNPs? Association mapping in plants, 2007: p. 41-52.
  11. Dakal, T.C., et al., Predicting the functional consequences of non-synonymous single nucleotide polymorphisms in IL8 gene. Scientific reports, 2017. 7(1): p. 6525.
  12. Collins, F.S., L.D. Brooks, and A. Chakravarti, A DNA polymorphism discovery resource for research on human genetic variation. Genome research, 1998. 8(12): p. 1229-1231.
  13. Mullaney, J.M., et al., Small insertions and deletions (INDELs) in human genomes. Human molecular genetics, 2010. 19(R2): p. R131-R136.
  14. Ramírez-Bello, J. and M. Jiménez-Morales, Functional implications of single nucleotide polymorphisms (SNPs) in protein-coding and non-coding RNA genes in multifactorial diseases. Gaceta medica de Mexico, 2017. 153(2): p. 238-250.
  15. Chauhan, W., et al., Cataloging the potential SNPs (single nucleotide polymorphisms) associated with quantitative traits, viz. BMI (body mass index), IQ (intelligence quotient) and BP (blood pressure): an updated review. Egyptian Journal of Medical Human Genetics, 2022. 23(1): p. 57.
  16. Shastry, B.S., SNPs in disease gene mapping, medicinal drug development and evolution. Journal of human genetics, 2007. 52: p. 871-880.
  17. Chen, R., et al., Non-synonymous and synonymous coding SNPs show similar likelihood and effect size of human disease association. PloS one, 2010. 5(10): p. e13574.
  18. Emilien, G., et al., Impact of genomics on drug discovery and clinical medicine. Qjm, 2000. 93(7): p. 391-423.
  19. Gonzaga-Jauregui, C., J.R. Lupski, and R.A. Gibbs, Human genome sequencing in health and disease. Annual review of medicine, 2012. 63: p. 35-61.
  20. Ramírez-Bello, J., G. Vargas-Alarcón, and J.M. Fragoso, Single nucleotide polymorphisms (SNPs): functional implications of regulatory-SNP (rSNP) and structural RNA (srSNPs) in complex diseases. Gaceta medica de Mexico, 2013. 149(2): p. 220-228.
  21. Sobieszczyk, M.E., J.R. Lingappa, and M.J. McElrath, Host genetic polymorphisms associated with innate immune factors and HIV-1. Current Opinion in HIV and AIDS, 2011. 6(5): p. 427-434.
  22. Sadee, W., et al., Pharmacogenomics of the RNA world: structural RNA polymorphisms in drug therapy. Clinical Pharmacology & Therapeutics, 2011. 89(3): p. 355-365.
  23. Sun, G., et al., SNPs in human miRNA genes affect biogenesis and function. Rna, 2009. 15(9): p. 1640-1651.
  24. Albert, F.W. and L. Kruglyak, The role of regulatory variation in complex traits and disease. Nature Reviews Genetics, 2015. 16(4): p. 197-212.
  25. Kwok, P.-Y. and X. Chen, Detection of single nucleotide polymorphisms. Current issues in molecular biology, 2003. 5(2): p. 43-60.
  26. Yue, P. and J. Moult, Identification and analysis of deleterious human SNPs. Journal of molecular biology, 2006. 356(5): p. 1263-1274.
  27. Oleykowski, C.A., et al., Mutation detection using a novel plant endonuclease. Nucleic acids research, 1998. 26(20): p. 4597-4602.
  28. Goldrick, M.M., RNase cleavage?based methods for mutation/SNP detection, past and present. Human Mutation, 2001. 18(3): p. 190-204.
  29. Sokurenko, E.V., et al., Detection of simple mutations and polymorphisms in large genomic regions. Nucleic Acids Research, 2001. 29(22): p. e111-e111.
  30. Orsini, L., et al., SNP discovery by mismatch-targeting of Mu transposition. Nucleic Acids Research, 2007. 35(6): p. e44.
  31. Janiak, A., et al., Application of degenerate oligonucleotide primed PCR (DOP-PCR) for SNP discovery in soybean. Euphytica, 2008. 162: p. 249-256.
  32. Xu, J.-Y., G.-B. Xu, and S.-L. Chen, A new method for SNP discovery. BioTechniques, 2009. 46(3): p. 201-208.
  33. Cotsapas, C. and D.A. Hafler, Immune-mediated disease genetics: the shared basis of pathogenesis. Trends in immunology, 2013. 34(1): p. 22-26.
  34. Wu, X., H. Chen, and H. Xu, The genomic landscape of human immune-mediated diseases. Journal of human genetics, 2015. 60(11): p. 675-681.
  35. Diaz-Gallo, L.-M. and J. Martin, Common genes in autoimmune diseases: a link between immune-mediated diseases. Expert Review of Clinical Immunology, 2012. 8(2): p. 107-109.
  36. Parkes, M., et al., Genetic insights into common pathways and complex relationships among immune-mediated diseases. Nature Reviews Genetics, 2013. 14(9): p. 661-673.
  37. Baune, B.T., et al., Interleukin-6 gene (IL-6): a possible role in brain morphology in the healthy adult brain. Journal of neuroinflammation, 2012. 9(1): p. 1-9.
  38. Fugger, L., L.T. Jensen, and J. Rossjohn, Challenges, progress, and prospects of developing therapies to treat autoimmune diseases. Cell, 2020. 181(1): p. 63-80.
  39. Wang, L., F.S. Wang, and M.E. Gershwin, Human autoimmune diseases: a comprehensive update. Journal of internal medicine, 2015. 278(4): p. 369-395.
  40. Selmi, C., Autoimmunity in 2019. Clinical Reviews in Allergy & Immunology, 2020. 59(3): p. 275-286.
  41. Chen, J.-Q., P. Szodoray, and M. Zeher, Toll-like receptor pathways in autoimmune diseases. Clinical reviews in allergy & immunology, 2016. 50: p. 1-17.
  42. Zeggini, E., et al., Translational genomics and precision medicine: Moving from the lab to the clinic. Science, 2019. 365(6460): p. 1409-1413.
  43. Cho, N.H., et al., IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes research and clinical practice, 2018. 138: p. 271-281.
  44. Atlas, D., IDF diabetes atlas. International Diabetes Federation (9th editio). Retrieved from http://www. idf. org/about-diabetes/facts-figures, 2019.
  45. Sladek, R., et al., A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature, 2007. 445(7130): p. 881-885.
  46. Scott, L.J., et al., A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. science, 2007. 316(5829): p. 1341-1345.
  47. Vatankhah Yazdi, K., et al., SLC30A8, CDKAL1, TCF7L2, KCNQ1 and IGF2BP2 are associated with type 2 diabetes mellitus in Iranian patients. Diabetes, Metabolic Syndrome and Obesity, 2020: p. 897-906.
  48. Jan, A., et al., Association of HLA-B Gene Polymorphisms with Type 2 Diabetes in Pashtun Ethnic Population of Khyber Pakhtunkhwa, Pakistan. Journal of Diabetes Research, 2021. 2021.
  49. Shi, Y., et al., Meta-analyses of the association of G6PC2 allele variants with elevated fasting glucose and type 2 diabetes. PLoS One, 2017. 12(7): p. e0181232.
  50. Hu, C., et al., A genetic variant of G6PC2 is associated with type 2 diabetes and fasting plasma glucose level in the Chinese population. Diabetologia, 2009. 52: p. 451-456.
  51. Li, C., et al., Glucose metabolism-related gene polymorphisms as the risk predictors of type 2 diabetes. Diabetology & Metabolic Syndrome, 2020. 12: p. 1-6.
  52. Muller, Y.L., et al., Common genetic variation in the glucokinase gene (GCK) is associated with type 2 diabetes and rates of carbohydrate oxidation and energy expenditure. Diabetologia, 2014. 57: p. 1382-1390.
  53. Qi, Q., et al., Association of GCKR rs780094, alone or in combination with GCK rs1799884, with type 2 diabetes and related traits in a Han Chinese population. Diabetologia, 2009. 52: p. 834-843.
  54. Sadeghi, M.B., et al., Significant association of LXR? (NR1H2) polymorphisms (rs28514894, rs2303044) with type 2 diabetes mellitus and laboratory characteristics. Journal of Diabetes & Metabolic Disorders, 2021. 20: p. 261-270.
  55. Sargazi, S., et al., SNPs in the 3?-untranslated region of SLC30A8 confer risk of type 2 diabetes mellitus in a south-east Iranian population: Evidences from case-control and bioinformatics studies. Journal of Diabetes & Metabolic Disorders, 2020. 19: p. 979-988.
  56. Ayelign, B., et al., TNF-? (? 308) Gene Polymorphism and Type 2 Diabetes Mellitus in Ethiopian Diabetes Patients. Diabetes, metabolic syndrome and obesity: targets and therapy, 2019: p. 2453-2459.
  57. Kallaur, A.P., et al., Genetic polymorphisms associated with the development and clinical course of multiple sclerosis. International journal of molecular medicine, 2011. 28(4): p. 467-479.
  58. Dominguez?Mozo, M., et al., Toll?like receptor?9 in S panish multiple sclerosis patients: an association with the gender. European Journal of Neurology, 2014. 21(3): p. 537-540.
  59. Ching, Y.M., et al., Association of CD58 polymorphism and multiple sclerosis in Malaysia: a pilot study. Autoimmunity Highlights, 2019. 10: p. 1-5.
  60. Hamid, M.M.A.E., et al., Multiple sclerosis: an associated single-nucleotide polymorphism study on Egyptian population. Network Modeling Analysis in Health Informatics and Bioinformatics, 2020. 9: p. 1-7.
  61. Esserlind, A.L., et al., Replication and meta?analysis of common variants identifies a genome?wide significant locus in migraine. European journal of neurology, 2013. 20(5): p. 765-772.
  62. Roshandel, D., et al., Genetic predisposition to ocular surface disorders and opportunities for gene-based therapies. The Ocular Surface, 2023.
  63. Tan, M.-S., et al., Genome-wide association studies in neurology. Annals of translational medicine, 2014. 2(12).
  64. Gormley, P., et al., Meta-analysis of 375,000 individuals identifies 38 susceptibility loci for migraine. Nature genetics, 2016. 48(8): p. 856-866.
  65. Kossowsky, J., et al., Association of genetic variants with migraine subclassified by clinical symptoms in adult females. Frontiers in Neurology, 2021. 11: p. 617472.
  66. Lin, Q.-F., et al., Association of genetic loci for migraine susceptibility in the she people of China. The journal of headache and pain, 2015. 16(1): p. 1-6.
  67. Kaur, S., et al., Role of single nucleotide polymorphisms (SNPs) in common migraine. The Egyptian Journal of Neurology, Psychiatry and Neurosurgery, 2019. 55(1): p. 1-7.
  68. Genome-wide association study of migraine implicates a common susceptibility variant on 8q22. 1. Nature genetics, 2010. 42(10): p. 869-873.
  69. Klimov, E., et al., Genetics of migraine-is there any progress. J Neurol Stroke, 2017. 7(4): p. 00245.
  70. van den Maagdenberg, A.M., et al., Migraine: gene mutations and functional consequences. Current opinion in neurology, 2007. 20(3): p. 299-305.
  71. Kumar, Y.S., et al., Association of RAMP-1 gene polymorphisms and risk of migraine: Systemic review and meta-analysis. Human Gene, 2023: p. 201157.
  72. Gorecki, A.M., et al., Single nucleotide polymorphisms associated with gut homeostasis influence risk and age-at-onset of Parkinson's disease. Frontiers in aging neuroscience, 2020. 12: p. 603849.
  73. Goldman, S.M., et al., Peptidoglycan recognition protein genes and risk of Parkinson's disease. Movement Disorders, 2014. 29(9): p. 1171-1180.
  74. Marras, C., C.G. Canning, and S.M. Goldman, Environment, lifestyle, and Parkinson's disease: implications for prevention in the next decade. Movement Disorders, 2019. 34(6): p. 801-811.
  75. Burgueño, J.F. and M.T. Abreu, Epithelial Toll-like receptors and their role in gut homeostasis and disease. Nature Reviews Gastroenterology & Hepatology, 2020. 17(5): p. 263-278.
  76. de Matos Lourenço, C., et al., Characterization and strong risk association of TLR2 del-196 to-174 polymorphism and Helicobacter pylori and their influence on mRNA expression in gastric cancer. World Journal of Gastrointestinal Oncology, 2020. 12(5): p. 535.
  77. Theunissen, F., et al., Structural variants may be a source of missing heritability in sALS. Frontiers in Neuroscience, 2020. 14: p. 47.
  78. Mann, D.M., Pyramidal nerve cell loss in Alzheimer's disease. Neurodegeneration, 1996. 5(4): p. 423-427.
  79. Tortora, F., et al., CD33 rs2455069 SNP: correlation with alzheimer’s disease and hypothesis of functional role. International Journal of Molecular Sciences, 2022. 23(7): p. 3629.
  80. Kunkle, B.W., et al., Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates A?, tau, immunity and lipid processing. Nature genetics, 2019. 51(3): p. 414-430.
  81. Wang, L., et al., Analysis of Genetic Association Between ABCA7 Polymorphism and Alzheimer’s Disease Risk in the Southern Chinese Population. Frontiers in Aging Neuroscience, 2022. 14.
  82. Halova, A., et al., CHAT gene polymorphism rs3810950 is associated with the risk of Alzheimer’s disease in the Czech population. Journal of biomedical science, 2018. 25: p. 1-9.
  83. Zhou, Q., et al., Association between APOC1 polymorphism and Alzheimer’s disease: a case-control study and meta-analysis. PloS one, 2014. 9(1): p. e87017.
  84. Karki, R., et al., Defining “mutation” and “polymorphism” in the era of personal genomics. BMC medical genomics, 2015. 8: p. 1-7.
  85. Zhang, H., et al., Meta-analysis of GWAS on both Chinese and European populations identifies GPR173 as a novel X chromosome susceptibility gene for SLE. Arthritis research & therapy, 2018. 20(1): p. 1-8.
  86. Zhang, Y., et al., Applications and potential of genome editing in crop improvement. Genome biology, 2018. 19: p. 1-11.
  87. Gosavi, G., et al., Applications of CRISPR technology in studying plant-pathogen interactions: overview and perspective. Phytopathology Research, 2020. 2(1): p. 1-9.

Photo
Kainat Ramzan
Corresponding author

Department of Biochemistry, University of Okara, Punjab, Pakistan

Photo
Ali Noman
Co-author

Department of Zoology, University of Okara, Punjab, Pakistan

Photo
Moeen Zulfiqar
Co-author

Department of Molecular Biology, Faculty of Life Sciences, University of Okara, Punjab, Pakistan

Photo
Saira Ramzan
Co-author

Department of Zoology, University of Okara, Punjab, Pakistan

Photo
Ayesha Waheed
Co-author

Department of Biochemistry, University of Okara, Punjab, Pakistan

Photo
Ali Haider Ali
Co-author

Department of Biochemistry, University of Okara, Punjab, Pakistan

Kainat Ramzan , Ali Noman , Saira Ramzan , Ali Haider Ali , Ayesha Waheed , Muhammad Bilal , Usama Tahir , Moeen Zulfiqar , Significance Of Genetic Polymorphisms In Autoimmune Diseases: A Comprehensive Review, Int. J. of Pharm. Sci., 2024, Vol 2, Issue 9, 399-412. https://doi.org/10.5281/zenodo.13731724

More related articles
A Review On Anti-Diabetic Effect Of Neem (Azadirac...
Yogita Dhuri , Vishal Biswas, Shamili Singh, Shruti Rathore, Divy...
Comprehensive Phytochemical Profiling And Therapeu...
Dr. Mallappa Shalavadi, Devraj Chavan, Shivaraj Hiremath, Vinayak...
A Systemic Review On Diuretic Activities Of Aquati...
G venkata Karthik Kumar Reddy , Shravya , Sinchana urs K N, Sneha...
A Comparative Study Evaluating The Level Of Urea And Creatinine In Males And Fem...
Zaman subhi madlool, Ahmed aziz, Dmoaa majid nasar, Doaa hazem mohammed , ...
Extraction, Phytochemical Investigation and Antimicrobial Activity of Plumeria o...
Ganesh D. Barkade, Akshata B. Sinare, Mayur G. Tandale, Rutuja B. Sonawane, Rutuja R. Sumbe, ...
A Review On The Analytical Method Development And Validation Of Sertralinetablet...
Ashishkumar Panpatil, Vedant Salunke , Snehal Jadhav, Faisal Shaikh, Pravin Jadhav, Vinod Bairagi, K...
Related Articles
Formulation And Evaluation of Momocardica Charantina Linn. Leaf Extracts Used to...
Dr. Khomendra Kumar Sarwa, Manisha chandrakar, Neha Sharma, Anjali Wahane, ...
Review A Mucoadhesive drug delivery system...
Rutuja S. Pawar, Sukumar M. Lande, Rupali R. Bendgude, ...
Formulation and Evaluation of Herbal Chocolate: Chocogil ...
Rajashri R. Kulkarni, Yogita Patil, Prajakta Kokane, ...
A Review on Pyranopyrazole as an Antibacterial Agent...
Wagh Shital Vilas , Dr. Prerana Jadhav, Dr. Pradyumna Ige, ...
A Review On Anti-Diabetic Effect Of Neem (Azadirachta Indica) Leaves...
Yogita Dhuri , Vishal Biswas, Shamili Singh, Shruti Rathore, Divyani Soni, ...
More related articles
A Review On Anti-Diabetic Effect Of Neem (Azadirachta Indica) Leaves...
Yogita Dhuri , Vishal Biswas, Shamili Singh, Shruti Rathore, Divyani Soni, ...
Comprehensive Phytochemical Profiling And Therapeutic Applications Of Plecosperm...
Dr. Mallappa Shalavadi, Devraj Chavan, Shivaraj Hiremath, Vinayaka Madivalar, Shubham Teli, ...
A Systemic Review On Diuretic Activities Of Aquatic Plants....
G venkata Karthik Kumar Reddy , Shravya , Sinchana urs K N, Sneha R V, Sneha Binu, Sonam Choedon, ...
A Review On Anti-Diabetic Effect Of Neem (Azadirachta Indica) Leaves...
Yogita Dhuri , Vishal Biswas, Shamili Singh, Shruti Rathore, Divyani Soni, ...
Comprehensive Phytochemical Profiling And Therapeutic Applications Of Plecosperm...
Dr. Mallappa Shalavadi, Devraj Chavan, Shivaraj Hiremath, Vinayaka Madivalar, Shubham Teli, ...
A Systemic Review On Diuretic Activities Of Aquatic Plants....
G venkata Karthik Kumar Reddy , Shravya , Sinchana urs K N, Sneha R V, Sneha Binu, Sonam Choedon, ...