Fact-checked by Grok 2 weeks ago

Genetic marker

A genetic marker is a DNA sequence with a known physical location on a chromosome, often exhibiting polymorphism, that serves as a reference point to track inheritance patterns and link specific genomic regions to traits, diseases, or ancestry. These markers typically consist of short segments of DNA that do not encode genes themselves but vary between individuals due to differences in nucleotide sequences, such as single nucleotide polymorphisms (SNPs) or insertions/deletions, enabling their use in distinguishing genetic variation across populations. By analyzing recombination frequencies during meiosis, genetic markers help construct linkage maps that reveal the relative positions of genes on chromosomes, facilitating the identification of disease-causing mutations. Genetic markers are fundamental tools in because they allow researchers to correlate phenotypic traits with underlying genetic factors without directly sequencing entire genomes, which was particularly valuable before high-throughput sequencing became widespread. Their , , and from environmental influences make them reliable for applications like paternity testing, forensic analysis, and studies. For instance, markers have been instrumental in positional cloning, where they narrow down genomic intervals containing disease genes by tracking co-inheritance with affected phenotypes in families. Common types of genetic markers include single nucleotide polymorphisms (SNPs), which are single base-pair variations occurring approximately every 300 bases in the and are widely used due to their abundance and ease of ; microsatellites (short tandem repeats), consisting of repeating units of 1-6 base pairs that exhibit high polymorphism and are useful for linkage analysis; and restriction fragment length polymorphisms (RFLPs), early markers based on variations in DNA cutting sites recognized by restriction enzymes. Other types encompass amplified fragment length polymorphisms (AFLPs) for rapid screening of multiple loci and insertion/deletion polymorphisms (indels) for tracking structural variations. The choice of marker type depends on factors like resolution needed, cost, and the studied, with SNPs now predominant in large-scale genome-wide association studies (GWAS) owing to their scalability. Applications of genetic markers span , , and , including mapping quantitative trait loci (QTLs) to identify genes influencing like or disease susceptibility, and enabling in breeding programs to accelerate the development of improved and varieties. In human health, they support diagnostic tests for inherited disorders, such as , by detecting specific genetic variants in disease-associated genes. Additionally, inform to predict drug responses based on genetic profiles. Ancestry-informative markers help trace patterns by revealing population-specific frequencies. Advances in sequencing technologies continue to expand their utility, integrating markers with whole- data for precise approaches.

Fundamentals

Definition and Characteristics

A genetic marker is a specific DNA sequence or gene with a known physical location on a chromosome, serving as a point of variation that enables the identification of particular genomic regions or sequence differences associated with traits, diseases, or ancestry. These markers are typically polymorphic, meaning they exhibit variations in nucleotide sequences among individuals within a population, allowing differentiation based on genetic diversity. Unlike genes, which encode functional products such as proteins, genetic markers often do not code for proteins but instead act as identifiable landmarks in the genome for tracking inheritance patterns. Key characteristics of genetic markers include their polymorphism, which provides the basis for distinguishing genetic variants; , as they are stably transmitted from parents to offspring following Mendelian principles; and linkage to traits of interest, where markers located near functional genes can co-segregate with those traits across generations. They demonstrate stability over generations due to the fidelity of and transmission, with minimal mutation rates in non-coding regions, and are selected for ease of detection through various molecular assays. For instance, single nucleotide polymorphisms (SNPs) exemplify polymorphic sites where a single base difference creates detectable variation. In linkage analysis, genetic markers play a central role by revealing the chromosomal positions of disease-associated genes through observed patterns of co-inheritance in families, facilitating without directly observing the causal variant. At the population level, concepts like and heterozygosity underpin the utility of genetic markers in assessing variation. quantifies the prevalence of a specific at a marker locus, calculated as the proportion of that among all alleles in the ; for a biallelic locus, the frequency p of one is given by p = \frac{\text{number of copies of the [allele](/page/Allele)}}{\text{total number of alleles sampled}} where the total alleles equal twice the number of individuals if diploid. This metric reveals -level differences in , with deviations from expected frequencies indicating evolutionary forces like selection or drift. Heterozygosity, the proportion of individuals carrying two different s at a locus, measures the degree of polymorphism and at that marker, often correlating with higher informativeness for linkage studies.

Historical Development

The foundations of genetic markers trace back to the mid-19th century with Gregor Mendel's experiments on pea plants, where he established the laws of and independent assortment—demonstrating that traits are transmitted as discrete units, laying the groundwork for identifying heritable variations as markers. In the early 20th century, phenotypic markers emerged, such as the discovered by in 1901, which identified heritable differences in human red blood cells based on reactions, enabling early applications in and paternity testing. However, these early markers were limited to observable traits and struggled to map complex or invisible genetic variations, restricting their utility in detailed analysis. The shift to molecular genetic markers began in the late 1970s with the development of restriction fragment length polymorphisms (RFLPs), introduced by David Botstein and colleagues in 1980, who proposed using restriction enzymes to detect DNA sequence variations as polymorphic sites for constructing maps in humans. This innovation allowed for the identification of differences as markers, overcoming the limitations of phenotypic approaches by enabling genome-wide mapping without relying on visible traits. Complementing this, conceived the () in 1983, a technique for amplifying specific DNA segments exponentially, which revolutionized marker detection by facilitating the analysis of minute genetic samples and integrating seamlessly with RFLP methods. In the 1980s, further advances came with the discovery of minisatellites—highly variable sequences—by in 1984, leading to the invention of DNA fingerprinting, a technique that used these markers for individual identification in forensics and paternity cases due to their unique, hypervariable patterns across genomes. The (HGP), launched in 1990 and completed in 2003, markedly accelerated the use of genetic markers by generating high-density maps of RFLPs, microsatellites, and other variants, which supported the sequencing of the entire human genome and identified millions of potential marker sites. Post-HGP, the focus shifted to single nucleotide polymorphisms (SNPs) as more precise markers, with the releasing its Phase I dataset in 2005, cataloging over 1.1 million SNPs across diverse populations to reveal haplotype structures and facilitate association studies for disease genes. Building on this, the (2008–2015) provided a deeper catalog of by sequencing the genomes of over 2,500 individuals from various populations, identifying more than 88 million variants, including 84 million SNPs, which expanded the repertoire of genetic markers available for research into rare variants and structural variations. In the 2020s, genetic markers have integrated with CRISPR-Cas9 genome editing, enabling targeted modification of marker sites for functional studies and therapeutic interventions, such as repairing disease-associated variants in model organisms and human cells.

Classification

Molecular Markers

Molecular markers are DNA-based genetic variations that occur at the sequence level, providing stable and heritable indicators of . These markers arise primarily from mutations, replication errors, and recombination events during and , enabling their use in identifying polymorphisms across genomes. Unlike phenotypic traits, molecular markers directly reflect nucleotide-level changes and are typically inherited in a co-dominant manner, allowing both alleles to be detected in heterozygous individuals. The most prevalent subtype is single nucleotide polymorphisms (SNPs), which involve a single base substitution at a specific position in the DNA sequence, such as an A to G transition. SNPs are biallelic, meaning they typically have two possible variants (e.g., C/T), and they constitute the vast majority of human genetic variation, with over 99.9% of detected variants in a typical genome being SNPs or short indels. For instance, SNPs in the BRCA1 gene, such as rs16941 (E1038G), have been associated with increased breast cancer risk, particularly in interaction with environmental factors like smoking in premenopausal women or hormone therapy in postmenopausal women. Microsatellites, also known as short tandem repeats (STRs), consist of tandemly repeated units of 1-6 base pairs, such as the dinucleotide motif (CA)_n, where n varies in length between individuals, leading to high polymorphism. These repeats arise mainly from slipped strand mispairing during , resulting in expansion or contraction of the repeat array. Microsatellites exhibit mutation rates around 10^{-3} per locus per generation, orders of magnitude higher than unique sequence DNA, which contributes to their utility in forensics for individual identification due to this variability. Insertions/deletions (indels) represent another subtype, involving the addition or removal of small segments, often 1-50 pairs, which can disrupt reading frames or alter protein function. Indels typically originate from replication slippage or errors in processes. Copy number variations (CNVs) encompass larger-scale duplications or deletions affecting thousands of pairs or more, generated through mechanisms like non-allelic (NAHR) or fork stalling and template switching during replication. Both indels and CNVs contribute to structural diversity but are less frequent than SNPs, impacting and expression. Overall, these molecular markers follow co-dominant patterns, where both maternal and paternal alleles are equally expressed and detectable, facilitating precise and without dominance effects obscuring heterozygotes.

Biochemical Markers

Biochemical genetic markers primarily involve variations at the protein level, such as protein polymorphisms arising from substitutions that alter protein function or structure. These markers, often detected through techniques like , include allozymes, which are variant forms of enzymes differing in electrophoretic mobility due to changes in their sequences. In the , protein revolutionized the study of by revealing extensive polymorphisms in natural populations, with early work demonstrating that approximately one-third of genes in humans were polymorphic based on protein s. , a subset of this approach, allowed for the identification of codominant patterns in proteins, facilitating early studies before the widespread use of DNA-based methods.

Detection Techniques

Traditional Methods

Traditional methods for detecting genetic markers relied on low-throughput, gel-based laboratory techniques developed primarily in the 1970s and 1980s, which exploited variations in DNA sequence to produce observable differences in fragment patterns. These approaches, such as (RFLP) and Southern blotting, formed the foundation for early genetic mapping and analysis before the advent of and sequencing technologies. They typically involved enzymatic digestion of DNA, separation by , and hybridization to identify polymorphisms, offering co-dominant markers useful for linkage studies but demanding significant hands-on effort. One of the earliest and most influential techniques was RFLP, introduced as a means to construct maps by detecting sequence variations that alter restriction sites. In RFLP, genomic DNA is digested with restriction endonucleases like , which recognize specific sequences (e.g., GAATTC) and cleave DNA at those sites, producing fragments of varying lengths due to polymorphisms such as single nucleotide polymorphisms (SNPs) or insertions/deletions (INDELs). The protocol for RFLP analysis includes the following steps:
  1. Extract and purify high-quality genomic DNA from the sample.
  2. Digest the DNA with a such as or in a at 37°C for several hours.
  3. Separate the resulting fragments by size using , where smaller fragments migrate faster.
  4. Transfer the separated DNA to a or membrane via Southern blotting.
  5. Hybridize the membrane with a labeled DNA probe complementary to the target sequence, followed by detection via autoradiography or to visualize polymorphic bands.
RFLP markers are co-dominant, allowing distinction between homozygous and heterozygous states, and were pivotal in early mapping efforts, with Botstein et al. estimating about 150 such markers would suffice for a comprehensive linkage spaced at 0.2 Morgans. Advantages include high specificity and applicability to forensics and disease diagnostics, but limitations encompass labor-intensive DNA isolation, the need for prior knowledge to design probes, and low throughput due to reliance on gel-based visualization. Amplified fragment length polymorphism (AFLP) emerged in the mid-1990s as a hybrid method combining restriction digestion with (PCR) to enhance sensitivity and generate multiple markers simultaneously. In AFLP, genomic DNA is first digested with restriction enzymes like and MseI, followed by of synthetic adapters to the fragment ends; selective PCR then amplifies a subset of these fragments using primers with degenerate bases for specificity. The amplified products are separated by or and scored for presence/absence of bands, revealing polymorphisms without requiring prior genomic sequence information. This technique, detailed by Vos et al., offers advantages such as high reproducibility, minimal DNA requirements (as low as 50-100 ng), and utility in studies, though it remains labor-intensive for primer optimization and produces dominant markers that cannot easily distinguish heterozygotes. Randomly amplified polymorphic DNA (RAPD) provided a simpler, PCR-based for marker screening, utilizing short arbitrary primers (typically 10 ) to amplify random DNA segments under low-stringency conditions. Developed by Welsh and McClelland in 1990, RAPD involves denaturing DNA, annealing the primer to multiple sites, and extending via to produce a pattern of bands visualized on gels after ; polymorphic bands arise from sequence variations affecting primer binding or amplification efficiency. This method was particularly valuable in early for assessing and constructing linkage maps, as it requires no prior sequence knowledge and can be completed in a single day with minimal equipment. However, RAPD's dominant nature, sensitivity to reaction conditions, and potential for non-reproducible results limit its reliability compared to more controlled techniques. Southern blotting served as a core visualization and detection step across these methods, particularly in RFLP, by transferring electrophoresed DNA fragments to a for hybridization with labeled probes specific to the genetic marker of interest. The process, pioneered by in 1975, involves alkali denaturation of gel-separated DNA, capillary transfer to the , and incubation with a radiolabeled or enzymatically tagged probe that binds complementary sequences, enabling detection of specific fragments as dark bands on film. This hybridization step confirms marker identity and size, providing quantitative insights into gene copy number or rearrangements, such as oncogene amplifications in cancer. Overall, these traditional protocols were cost-effective for small-scale studies but constrained by their manual nature and dependence on sequence-specific probes.

Modern and Advanced Methods

Modern and advanced methods for detecting genetic markers leverage high-throughput sequencing and computational tools to enable scalable, precise analysis of genetic variations such as single nucleotide polymorphisms (SNPs), copy number variations (CNVs), and epigenetic modifications. These techniques have revolutionized marker identification by processing vast datasets rapidly and cost-effectively, facilitating genome-wide studies that were previously infeasible. Next-generation sequencing (NGS) platforms, such as Illumina's NovaSeq, are widely used for and whole-genome sequencing (WGS), allowing simultaneous interrogation of millions of markers across samples. By 2023, the cost of WGS had decreased to approximately $600 per sample, driven by improvements in sequencing throughput and efficiency, making it accessible for large-scale studies. As of 2025, costs have further declined to around $100–$200 per , enhancing scalability for population-level marker . , an extension of NGS, resolves marker heterogeneity within tissues by profiling individual cells, revealing cell-type-specific variations in SNPs and epigenetic markers that bulk sequencing overlooks. For instance, integrating single-cell sequencing with has identified genetic drivers of heterogeneity in complex traits like . Advanced tools further enhance marker detection and validation. CRISPR-Cas9 enables precise editing and correction of SNPs, with protocols developed around 2022 demonstrating high-efficiency base editing in induced pluripotent stem cells to model and validate pathogenic markers without off-target effects. Computational methods complement these experimental approaches, particularly in genome-wide association studies (GWAS). Software like PLINK performs marker-trait association analysis by calculating (LD), a measure of non-random associations at nearby loci. LD is quantified using the coefficient D' = \frac{D}{D_{\max}}, where D = p_{AB} - p_A p_B is the disequilibrium, and D_{\max} = \min(p_A p_B, q_A q_B) if D > 0 (or the appropriate minimum for D < 0, with q_A = 1 - p_A, q_B = 1 - p_B). To derive this, compute D first; then normalize by the theoretical maximum D possible under observed frequencies to scale D' between -1 and 1, standardizing LD strength. PLINK assesses LD significance via a test, where the statistic \chi^2 = n D^2 / (p_A (1 - p_A) p_B (1 - p_B)) (approximating n r^2, with n as sample size) follows a chi-square distribution with 1 degree of freedom under the of no LD, enabling computation for marker associations. Recent innovations integrate (AI) with these methods for enhanced prediction. AlphaFold3, released in 2024, models protein structures and interactions with high accuracy, aiding the prediction of epigenetic markers. Additionally, single-molecule real-time (SMRT) sequencing via PacBio platforms excels in long-read CNV detection, resolving complex structural variations that short-read NGS misses, such as tandem amplifications underlying genetic disorders. These long reads, often exceeding 10 kb, provide phased haplotypes and direct epigenetic readouts, improving marker resolution in clinical .

Applications

In Genetic Research and Mapping

Genetic markers serve as essential tools in genetic research by facilitating the construction of linkage maps, which identify the relative positions of genes and loci on chromosomes based on recombination frequencies during . These maps rely on polymorphic markers, such as restriction fragment length polymorphisms (RFLPs) or single nucleotide polymorphisms (SNPs), that co-segregate with target genes in mapping populations derived from crosses between genetically diverse individuals. By analyzing inheritance patterns, researchers can estimate genetic distances in , where 1 cM corresponds to a 1% recombination rate. A key statistical method in linkage analysis is the logarithm of odds (LOD) score, which quantifies the likelihood of linkage between a marker and a trait locus versus assortment. The LOD score is calculated as \text{LOD}(\theta) = \log_{10} \left( \frac{[L](/page/Fraction)(\theta)}{[L](/page/Fraction)(0.5)} \right), where L(\theta) is the likelihood of the observed data given a recombination \theta (the probability of recombination between loci, ranging from 0 to 0.5), and L(0.5) assumes no linkage ( at \theta = 0.5). To compute this, one first determines the pedigree's ( configuration) and counts recombinant versus non-recombinant offspring; the likelihood L(\theta) is then derived from the multinomial probability of these counts under the assumed \theta, often maximized via expectation-maximization algorithms for multipoint analysis across multiple markers. A LOD score greater than 3 typically indicates significant linkage, as it corresponds to of at least 1000:1 against the , enabling fine-scale mapping of genes. In quantitative trait locus (QTL) mapping, genetic markers are used to detect genomic regions contributing to complex, polygenic traits influenced by multiple genes and environmental factors. This involves creating a segregating (e.g., or recombinant inbred lines) from parental strains differing in the trait, genotyping with markers like SNPs or simple sequence repeats (SSRs), and performing statistical tests such as interval mapping or composite interval mapping to identify marker-trait associations. For instance, peaks in LOD score profiles along the indicate QTL positions, with effect sizes estimated from the proportion of phenotypic variance explained. A classic example is the identification of six QTLs for flowering time in , where the top five loci accounted for 84% of the variation, demonstrating how markers enable dissection of quantitative traits like or response. Genetic markers also underpin phylogenetic studies by reconstructing evolutionary relationships through marker-based trees, particularly using SNPs to infer population histories and ancestry. In the , over 88 million SNPs from 2,504 individuals across 26 populations were cataloged, allowing construction of haplotype phylogenies that reveal fine-scale patterns and events without relying on whole-genome alignments. Specific historical examples include the 1989 RFLP linkage map of , which integrated 94 markers (including cloned genes and cosmids) across five chromosomes from crosses between and Landsberg erecta ecotypes, providing a foundational framework for the plant's genome sequencing. Similarly, the genotyped over 1 million SNPs in 270 individuals from four populations to delineate haplotype blocks—regions of low recombination where alleles are inherited together—reducing the need for exhaustive genotyping and accelerating association studies. Beyond basic mapping, genetic markers drive (MAS) in agricultural research to breed crops with enhanced traits, such as . In , SSR markers linked to stay-green QTLs (e.g., Stg1 on SBI-03, explaining 20% phenotypic variance) have been used to introgress alleles into elite varieties, improving grain yield under water-limited conditions by selecting progeny with favorable marker haplotypes early in breeding cycles. This approach, applied in programs like those pyramiding Stg3 and Stg4 QTLs, has accelerated development of resilient hybrids without extensive phenotyping.

In Medicine and Forensics

Genetic markers play a pivotal role in by identifying susceptibility and guiding therapeutic decisions. Genome-wide association studies (GWAS) have linked specific single polymorphisms (SNPs) in the APOE gene, particularly the ε4 allele, to increased risk of late-onset , with carriers facing a 3- to 15-fold higher lifetime risk compared to non-carriers. In , variants in the gene influence and response; for instance, poor metabolizers may experience reduced or heightened from , as it converts poorly to active . The Clinical Pharmacogenetics Implementation Consortium (CPIC) provides guidelines for integrating genotyping into prescribing, with updates in 2023 emphasizing dose adjustments for antidepressants like to mitigate adverse effects in intermediate or poor metabolizers. In , mutations in and genes serve as key genetic markers for hereditary and risk. The BRCA1 185delAG founder mutation, prevalent in approximately 1% of Ashkenazi Jewish individuals, confers a 50-80% lifetime risk and is routinely tested in high-risk populations to inform preventive strategies such as enhanced surveillance or prophylactic surgery. arrays enable rapid, high-throughput detection of these and other SNPs, facilitating personalized diagnostics in clinical settings by analyzing thousands of markers simultaneously for conditions like or pharmacogenomic profiling. Forensic applications leverage genetic markers for human identification and kinship analysis. Short tandem repeat (STR) profiling, using the 20 core loci defined by the FBI's Combined DNA Index System (CODIS) since 2017, allows matching of crime scene DNA to suspects with extremely high specificity, and has aided over 751,000 investigations as of September 2025 through the national database. Mitochondrial DNA (mtDNA) markers, inherited solely through the maternal line, are valuable when nuclear DNA is degraded, as in old remains, enabling lineage tracing and exclusion of paternal contributors in cases like mass disasters. Paternity and kinship testing commonly employs 15 or more autosomal markers, achieving a probability of paternity exceeding 99.99% when the alleged matches the , providing court-admissible in legal disputes.

Emerging and Future Uses

In , genetic markers are increasingly targeted through advanced therapies like CRISPR-based editing, enabling precise modifications to disease-associated variants. For instance, the 2023 FDA approval of Casgevy, an autologous CRISPR-Cas9 gene-edited therapy, targets the BCL11A genetic marker to treat by reactivating production in patients aged 12 and older with recurrent vaso-occlusive crises. This milestone represents a shift toward marker-specific interventions in clinical practice, with ongoing trials exploring similar edits for other hemoglobinopathies and beyond. Artificial intelligence and big data analytics are revolutionizing genetic marker discovery by leveraging machine learning to predict epigenetic modifications from genomic sequences. Google DeepMind's AlphaGenome model, released in 2025, uses to forecast functional impacts of DNA variants on gene regulation, including epigenetic markers like chromatin accessibility, across sequences up to 1 million base pairs long. Such tools enhance the identification of non-coding regulatory markers, accelerating discoveries in complex traits and diseases by integrating vast datasets from projects like the . In environmental genomics, genetic markers facilitate biodiversity tracking through environmental DNA (eDNA) approaches, allowing non-invasive monitoring of species diversity in ecosystems. Post-2020 advancements in eDNA metabarcoding use targeted genetic markers, such as cytochrome c oxidase I (COI), to detect multiple taxa from water or soil samples, enabling large-scale assessments of biodiversity loss amid climate change. Similarly, eDNA metagenomics sequences entire microbial and eukaryotic communities, revealing marker-based shifts in ecosystem health for conservation efforts. Synthetic biology is advancing the design of novel genetic markers for applications like gene drives, which propagate engineered traits through populations. Recent CRISPR-based constructs for malaria vector control, such as those engineering resilient gene drives developed in 2024, predict and overcome target site resistance while incorporating synthetic markers to track spread in laboratory and field simulations of mosquitoes. These engineered markers, often fluorescent or resistance-linked, enable precise tracking and containment of gene drive spread in laboratory and field simulations. Integration of genetic markers with single-cell sequencing (scRNA-seq) has improved cancer subtyping by resolving intratumor heterogeneity post-2021. For example, combined scRNA-seq and analyses of pancreatic ductal adenocarcinoma samples have identified marker-defined subtypes based on transcriptional profiles of malignant cells, revealing distinct prognostic trajectories. In , scRNA-seq has pinpointed epigenetic and mutational markers in cellular states, aiding the classification of therapy-resistant populations. Looking ahead, real-time monitoring of genetic markers via wearables holds promise for proactive health management, with genetically programmable devices detecting fluctuations noninvasively. Emerging prototypes, such as microfluidic wearables integrated with diagnostics, could enable continuous tracking of circulating genetic variants from sweat or interstitial fluid, potentially alerting users to disease onset. However, ethical challenges arise in handling marker data from large databases, including privacy risks under evolving regulations like the EU's GDPR, which in 2025 strengthened safeguards for genetic information transfers in cross-border research through enhanced consent and adequacy decisions. These considerations underscore the need for robust frameworks to balance innovation with data protection in marker-driven applications.

References

  1. [1]
    Genetic Marker - National Human Genome Research Institute
    A genetic marker is a DNA sequence with a known physical location on a chromosome. Genetic markers can help link an inherited disease with the responsible gene.
  2. [2]
    Genetic marker: a genome mapping tool to decode genetic diversity ...
    DNA markers are sets of nucleotide bases of DNA sequence that exhibit polymorphism to differentiate individuals of a population (Jiang, 2013). DNA markers have ...
  3. [3]
    Genetic Mapping Fact Sheet
    Aug 17, 2020 · What are genetic markers? Markers themselves usually consist of DNA that does not contain a gene. But because markers can help a researcher ...
  4. [4]
    Genetics, Mutations, and Polymorphisms - NCBI - NIH
    Genetic markers are specific DNA sequences that are polymorphic and employed in mapping of disease genes. When a single disorder can be caused by mutation in ...
  5. [5]
    chapter 4-positional cloning of genetic disorders - PubMed
    The genetic markers define an interval that is a function of their recombination frequencies with the disease, in which the disease gene is localised.
  6. [6]
    Basic concepts and methodologies of DNA marker systems in plant ...
    The concepts, methodologies and applications of some of the major molecular or DNA markers commonly used in plant science have been presented.
  7. [7]
    A review on SNP and other types of molecular markers and their use ...
    Amongst others, the microsatellite DNA marker has been the most widely used, due to its easy use by simple PCR, followed by a denaturing gel electrophoresis ...Missing: applications | Show results with:applications
  8. [8]
    Application of linked DNA markers to screening families ... - PubMed
    In this report we discuss the use of these markers for early and accurate prediction of gene carrier status in three different families. Factors that influence ...
  9. [9]
    Ancestry-informative Markers
    Ancestry-informative markers are sets of polymorphisms for a particular DNA sequence that appear in substantially different frequencies between populations.
  10. [10]
    An Overview: Genetic Tumor Markers for Early Detection and ...
    We can detect the types of genes that are either up or down-regulated in specific cancer types with the use of microarray, real-time PCR, and digital PCR.<|control11|><|separator|>
  11. [11]
    Genetic markers - Latest research and news - Nature
    Genetic markers are DNA sequences with known physical locations on chromosomes. They are points of variation that can be used to identify individuals or species ...
  12. [12]
    Are Genes Good Markers of Biological Traits? - NCBI - NIH
    A protein that is consistently a symptom of the disease could not serve as a predictive marker, even though it would be present in all carriers of the disease ...
  13. [13]
    Forensic genetics - PMC - PubMed Central - NIH
    Genetic markers generally have features such as strong polymorphisms, codominant expression and ease of observation and recording.<|control11|><|separator|>
  14. [14]
    Genetic linkage analysis - PubMed - NIH
    Genetic linkage analysis is a powerful tool to detect the chromosomal location of disease genes. It is based on the observation that genes that reside ...
  15. [15]
    Genetic linkage analysis in the age of whole-genome sequencing
    For many years, linkage analysis was the primary tool used for the genetic mapping of Mendelian and complex traits with familial aggregation.
  16. [16]
    allele frequency | Learn Science at Scitable - Nature
    An allele frequency is calculated by dividing the number of times the allele of interest is observed in a population by the total number of copies of all the ...
  17. [17]
    A new analysis tool for individual-level allele frequency for genomic ...
    Allele frequency denotes the relative frequency of an allele compared with the total frequency of all alleles at a marker locus. It is one of the most important ...
  18. [18]
    Genetic variance components and heritability of multiallelic ...
    Jul 15, 2015 · Heritability of heterozygosity is highest for highly unequal allele frequencies and is reduced by inbreeding. Reductions in heritability with ...Biallelic Loci And No... · Multiallelic Loci And No... · Outbred PopulationsMissing: characteristics | Show results with:characteristics
  19. [19]
  20. [20]
    A Brief History of Human Blood Groups - PMC - NIH
    Landsteiner in his 17th scientific paper in 1901 reported blood group ABO which was displayed at the beginning with the letters ABC. In 1930, he received the ...
  21. [21]
    Construction of a genetic linkage map in man using restriction ...
    The method uses DNA probes to detect polymorphisms, defining loci. These loci are tested for linkage in pedigrees to form a genetic map.
  22. [22]
    Kary B. Mullis – Facts - NobelPrize.org
    Kary Mullis invented the process known as polymerase chain reaction (PCR), in which a small amount of DNA can be copied in large quantities over a short period ...
  23. [23]
    Hypervariable 'minisatellite' regions in human DNA - Nature
    Mar 7, 1985 · Hypervariable 'minisatellite' regions in human DNA. Alec J. Jeffreys,; Victoria Wilson &; Swee Lay Thein. Nature volume 314, pages 67–73 ...Abstract · Author Information · Authors And Affiliations<|separator|>
  24. [24]
    Human Genome Project Fact Sheet
    Jun 13, 2024 · In 2003, the Human Genome Project produced a genome sequence that accounted for over 90% of the human genome. It was as close to complete as the ...
  25. [25]
    International HapMap Project
    May 1, 2012 · The haplotype map, or "HapMap," is a tool that allows researchers to find genes and genetic variations that affect health and disease.
  26. [26]
    Integrating CRISPR Technology with Key Genetic Markers in ... - MDPI
    This article discusses how CRISPR Cas9 technology can be combined with these markers to create treatments. CRISPR allows for the alteration or repair of these ...
  27. [27]
    A global reference for human genetic variation - Nature
    ### Summary of SNPs Prevalence in Human Genetic Variation from the 1000 Genomes Project
  28. [28]
    BRCA1 polymorphisms and breast cancer epidemiology in the ... - NIH
    Some studies have reported associations between BRCA1 SNPs and both familial and sporadic breast cancer risk; however there is a lack of consistency across ...
  29. [29]
    A Brief Review of Short Tandem Repeat Mutation - PMC - NIH
    High Mutation Rates of STRs. Unique DNA sequences in a genome exhibit a very low mutation rate (approximately 10−9 nt per generation), whereas the mutation ...
  30. [30]
    DNA copy number variation: Main characteristics, evolutionary ... - NIH
    CNVs emerge from different mutational mechanisms, including DNA recombination-, replication- and repair-associated processes. Mechanisms of change in gene copy ...
  31. [31]
    Genetic Variation in Natural Populations - Oxford Academic
    Differences in amino acid sequence that alter the electrophoretic mobility of proteins, termed allozymes, were widely used to measure genetic variation and ...
  32. [32]
    The Electrophoretic Revolution in the 1960s - NIH
    By the beginning of the 1960s, the inception of zone electrophoresis in laboratories around the world transformed—some say, revolutionized—the study of proteins ...Missing: markers polymorphisms
  33. [33]
    Protein Polymorphism - an overview | ScienceDirect Topics
    Using this technique it was shown for the first time in 1966 that about a third of all the genes in chromosomes are polymorphic, and that the average individual ...
  34. [34]
    5 Perspectives of molecular genetics and fisheries into the 21st ...
    A successful search for Mendelian markers in the 1960s. Protein electrophoresis unveiled large amounts of previously unsuspected Mendelian variation in most ...
  35. [35]
    Genetics, Epigenetic Mechanism - StatPearls - NCBI Bookshelf - NIH
    Three different epigenetic mechanisms have been identified: DNA methylation, histone modification, and non-coding RNA (ncRNA)-associated gene silencing.
  36. [36]
    DNA Methylation and Its Basic Function | Neuropsychopharmacology
    Jul 11, 2012 · The methylation of CpG islands results in stable silencing of gene expression (Mohn et al, 2008). During gametogenesis and early embryonic ...
  37. [37]
    Epigenetic inheritance and the missing heritability - Human Genomics
    Jul 28, 2015 · Epigenetic modifications include DNA methylation and histone modifications. These have the ability to alter gene expression patterns without ...
  38. [38]
    The Impact of Nutrition and Environmental Epigenetics on Human ...
    Nov 1, 2018 · This review explores how the environment affects the epigenome in health and disease, with a particular focus on cancer.
  39. [39]
    Epigenetics in Prader-Willi Syndrome - PMC - PubMed Central
    PWS is both a genetic and epigenetic disorder, mapping the imprinted chromosomal domain of 15q11.2-13.3. Common to all cases of PWS is the absence of an ...
  40. [40]
    Restriction Fragment Length Polymorphism (RFLP) - NCBI - NIH
    Nov 9, 2017 · An RFLP probe is a labeled DNA sequence that hybridizes with one or more fragments of the digested DNA sample after they were separated by gel ...
  41. [41]
    Gene Analysis: DNA - Holland-Frei Cancer Medicine - NCBI Bookshelf
    The final goal of Southern blotting is to identify specific fragments of cut DNA by using nucleic acid hybridization. Because the agarose gel used in ...
  42. [42]
    Amplified Fragment Length Polymorphism (AFLP) - NIH
    AFLP is a PCR-based technique that uses selective amplification of a subset of digested DNA fragments to generate and compare unique fingerprints for genomes ...
  43. [43]
    Next-Generation Sequencing Technology: Current Trends and ... - NIH
    Next-generation sequencing (NGS) is a powerful tool used in genomics research. NGS can sequence millions of DNA fragments at once.
  44. [44]
    Advances in Whole Genome Sequencing: Methods, Tools, and ... - NIH
    Jan 4, 2025 · NGS is noted for its speed and cost-effectiveness, generating large volumes of sequencing data [44] and contributing significantly to various ...
  45. [45]
    Genome Wide Sampling Sequencing for SNP Genotyping - NIH
    It is true that customized SNP arrays/chips can be manufactured, but they are cost-prohibitive to many researchers. Next generation sequencing (NGS) ...
  46. [46]
    Leveraging illumina iSeq100 for whole genome sequencing of ...
    Sep 26, 2025 · This cost when filtered for India (closest to Nepal) was ~$55.5 to ~$173 per Gb. Furthermore, a systematic review of 2023 found the cost of WGS ...Methods · Library Preparation And... · Results
  47. [47]
    Genetic drivers of heterogeneity in type 2 diabetes pathophysiology
    Feb 19, 2024 · Our findings show the value of integrating multi-ancestry genome-wide association study data with single-cell epigenomics to disentangle the ...
  48. [48]
    CRISPR/Cas9 editing in iPSCs results in detrimental on-target ...
    The ability to precisely edit the genome of human induced pluripotent stem cell (iPSC) lines using CRISPR/Cas9 has enabled the devel-.
  49. [49]
    Advances in genomic tools for plant breeding - PubMed Central - NIH
    Nov 7, 2024 · More advanced techniques like serial analysis of gene expression (SAGE) and massively parallel signature sequencing (MPSS) address some of these ...<|separator|>
  50. [50]
    LD calculations - PLINK
    Jan 25, 2017 · PLINK includes a set of options to calculate pairwise linkage disequilibrium between SNPs, and to present or process this information in various ways.Missing: software GWAS marker-
  51. [51]
    Powerful new AI software maps virtually any protein interaction in ...
    May 8, 2024 · A free online platform that enables users to create AF3 models of proteins interacting with almost any other biomolecule.
  52. [52]
    Histone modification–driven structural remodeling unleashes ...
    Mar 26, 2025 · Our study suggests the mechanism by which conformational rearrangements in DNMT3B are triggered by histone modifications, ultimately unleashing its activity in ...Missing: marker | Show results with:marker
  53. [53]
    Long-read genome sequencing resolves complex genomic ... - Nature
    Dec 18, 2024 · Here we used Pacific Biosciences circular consensus sequencing to resolve complex rearrangements in two patients with rare genetic anomalies.
  54. [54]
    Approaches to long-read sequencing in a clinical setting to improve ...
    Oct 9, 2022 · Single-molecule long-read sequencing has the additional advantage of directly detecting epigenetic markers, typically methylated CpG ...
  55. [55]
    Statistical Approaches to Gene Mapping - PMC - NIH
    Allele frequency differences between the populations, for a given marker, are evidence for LD. LD may occur as a consequence of population admixture, but strong ...Genetic-Linkage Analysis · Disequilibrium Mapping · Microarray Expression DataMissing: characteristics | Show results with:characteristics
  56. [56]
    Quantitative Trait Locus (QTL) Analysis | Learn Science at Scitable
    ### Definition and Process of QTL Mapping Using Genetic Markers
  57. [57]
    Restriction Fragment Length Polymorphism Linkage Map ... - PubMed
    We have constructed a restriction fragment length polymorphism (RFLP) linkage map of the nuclear genome of the small flowering plant Arabidopsis thaliana.Missing: 1990s paper
  58. [58]
    1000 Genomes | A Deep Catalog of Human Genetic Variation
    ### Summary of 1000 Genomes Project Use of SNPs for Phylogenetics or Population Studies
  59. [59]
    Drought Tolerance and Application of Marker-Assisted Selection in ...
    Nov 30, 2021 · Marker-assisted selection is among the tools that are used for mapping QTLs conferring drought tolerance in sorghum. For instance, molecular ...
  60. [60]
    APOE and Alzheimer's Disease: Advances in Genetics ... - NIH
    The APOE ε4 allele remains the strongest genetic risk factor for sporadic Alzheimer's disease and the APOE ε2 allele the strongest genetic protective factor.
  61. [61]
    Clinical Pharmacogenetics Implementation Consortium (CPIC ... - NIH
    The purpose of this guideline is to provide information to allow the interpretation of clinical CYP2D6 genotype tests so that the results can be used to guide ...
  62. [62]
    Clinical Pharmacogenetics Implementation Consortium (CPIC ...
    Apr 9, 2023 · We provide recommendations for using CYP2D6, CYP2C19, and CYP2B6 genotype results to help inform prescribing these antidepressants and describe ...
  63. [63]
    The carrier frequency of the BRCA1 185delAG mutation is ... - Nature
    Oct 1, 1995 · Our results suggest that one in a hundred women of Ashkenazi descent may be at especially high risk of developing breast and/or ovarian cancer.
  64. [64]
    Array genotyping as diagnostic approach in medical genetics
    Aug 1, 2022 · Genotyping arrays are by far the most widely used genetic tests but are not generally utilized for diagnostic purposes in a medical context.
  65. [65]
    CODIS Archive — LE - FBI.gov
    ... Loci:The FBI Laboratory announced an expansion of the original 13 short tandem repeat (STR) loci that have been the core of NDIS since 1997. Seven additional ...
  66. [66]
    Mitochondrial DNA in forensic use - PMC - NIH
    Aug 10, 2021 · Mitochondrial DNA is considered to be inherited through the maternal line such that an individual's mtDNA will be shared with that person's ...
  67. [67]
    Understanding the Paternity Index & Probability of Paternity
    Oct 8, 2025 · Modern DNA testing is extraordinarily accurate when performed by accredited laboratories that follow strict quality standards. Labs use 15–25 ...
  68. [68]
    FDA Approves First Gene Therapies to Treat Patients with Sickle ...
    Dec 8, 2023 · The FDA approved the first cell-based gene therapies, Casgevy and Lyfgenia, for the treatment of sickle cell disease in patients 12 years ...
  69. [69]
    [PDF] December 8, 2023 Clinical Review Memo - CASGEVY - FDA
    Dec 8, 2023 · Sickle Cell Disease Treated with Autologous CRISPR-Cas9 Modified ... team recommends regular approval for exa-cel for the treatment of sickle cell.
  70. [70]
    [PDF] AlphaGenome: advancing regulatory variant effect prediction with a ...
    Deep learning models that predict functional genomic measurements from DNA sequence are powerful tools for deciphering the genetic regulatory code. Existing ...
  71. [71]
    AlphaGenome: AI for better understanding the genome
    Jun 25, 2025 · Our model analyzes up to 1 million DNA letters and makes predictions at the resolution of individual letters. Long sequence context is important ...Missing: epigenetic 2024
  72. [72]
    Unlocking natural history collections to improve eDNA reference ...
    Sep 11, 2025 · Environmental DNA (eDNA) metabarcoding offers a noninvasive method used for biodiversity monitoring and ecosystem assessment, but its accuracy ...
  73. [73]
    Improving Whole Biodiversity Monitoring and Discovery With ...
    Apr 1, 2025 · Environmental DNA (eDNA) metagenomics sequences all DNA molecules present in environmental samples and has the potential of identifying ...
  74. [74]
    Engineering Resilient Gene Drives Towards Sustainable Malaria ...
    Oct 21, 2024 · Synthetic gene drives can be used to spread desirable traits through natural pest or disease vector populations for sustainable genetic control.Results · Materials & Methods · Cloning Of Crispr Constructs...<|separator|>
  75. [75]
    Novel combination of CRISPR-based gene drives eliminates ...
    Mar 4, 2021 · We present HD-ClvR in this modelling study, a novel combination of CRISPR-based gene drives that eliminates resistance and localises spread.
  76. [76]
    Integrated single-cell and spatial transcriptomics uncover distinct ...
    Here, we perform single-cell/single-nucleus RNA sequencing (sc/snRNA-seq) and spatial transcriptomics on 62 samples from 25 pancreatic ductal adenocarcinoma ( ...
  77. [77]
    Single-cell RNA sequencing reveals different cellular states ... - Nature
    Aug 26, 2025 · Single-cell RNA sequencing (scRNA-seq) can reveal the distinct transcriptional profiles of individual malignant and non-malignant cells in the ...
  78. [78]
    Genetically programmable wearable devices for precision ... - PubMed
    Aug 4, 2025 · Wearable devices have emerged as powerful tools for continuous, real-time health monitoring, enabling the detection of biochemical markers ...
  79. [79]
    New German Guidelines on GDPR Requirements for International ...
    Oct 24, 2025 · ... GDPR, including any applicable exceptions for special categories of data such as health or genetic data. The second stage determines whether ...