Fact-checked by Grok 2 weeks ago

Cytogenetics

Cytogenetics is the branch of genetics that focuses on the study of chromosomes—their structure, function, behavior during cell division, inheritance patterns, and abnormalities—primarily through microscopic and molecular techniques to understand their role in heredity and disease. It encompasses the examination of chromosome number, size, shape, and banding patterns in cells from tissues such as blood, bone marrow, or amniotic fluid, often to detect anomalies like aneuploidy or structural rearrangements. This field bridges classical microscopy with modern genomics, enabling the diagnosis of genetic disorders and cancers by revealing how chromosomal variations contribute to phenotypic traits and pathologies. The history of cytogenetics traces back to the late , when improved staining techniques allowed visualization of thread-like structures in dividing cells, leading Wilhelm Waldeyer to coin the term "" in 1888. A pivotal milestone occurred in 1956, when Joe Hin Tjio and Albert Levan accurately determined the human number as 46, overturning earlier estimates and launching clinical cytogenetics as a discipline. Subsequent advancements, such as the use of to arrest cells in and hypotonic solutions for chromosome spreading in the , facilitated clearer imaging and led to landmark discoveries, including the identification of trisomy 21 in (1959) and the in chronic myeloid leukemia (1960). By the 1970s, banding techniques like enhanced resolution, allowing detection of microdeletions and translocations, while the 1980s introduced (FISH) for targeted gene mapping. Key techniques in cytogenetics include conventional karyotyping, which involves staining and photographing metaphase chromosomes to produce an arranged display (karyogram) for assessing gross abnormalities, and chromosomal microarray analysis (CMA) for higher-resolution detection of copy number variations without requiring cell culture. FISH uses fluorescent probes to bind specific DNA sequences, enabling rapid visualization of gene amplifications or deletions, while spectral karyotyping (SKY) and multicolor FISH (M-FISH) differentiate all chromosomes by color for complex rearrangements. These methods, often combined, provide resolutions from 5-10 megabases in traditional karyotypes to under 100 kilobases in arrays, supporting both constitutional (germline) and acquired (somatic) analyses. Applications of cytogenetics span prenatal diagnosis, where it detects fetal aneuploidies like via ; constitutional genetics for disorders such as (45,X); and for prognostic markers in and solid tumors. In cancer, cytogenetic profiling identifies driver mutations, such as BCR-ABL fusion in Philadelphia-positive leukemia, guiding targeted therapies. Integrations with next-generation sequencing have provided finer genomic insights, evolving cytogenetics into cytogenomics for , with recent advancements as of 2025 including optical genome mapping for improved structural variant detection and AI-assisted automated karyotyping.

Fundamentals

Definition and Scope

Cytogenetics is a branch of that focuses on the study of chromosomes, including their structure, number, inheritance, variation, and roles in heredity and . It examines how chromosomes carry genetic and influence phenotypic traits, serving as a foundational in understanding genomic organization and abnormalities. The scope of cytogenetics encompasses analysis of chromosomes in prenatal, postnatal, and cells, bridging classical techniques with approaches to detect heritable and acquired genetic variations. This interdisciplinary field applies to diagnosing congenital disorders, developmental delays, intellectual disabilities, and malignancies, such as hematologic and solid tumors, by identifying structural and numerical changes. It integrates cytological observations with genomic insights to inform clinical management and research in human and other eukaryotic organisms. Key concepts in cytogenetics include the chromosome as a DNA-protein complex, where DNA encodes genetic material and proteins like histones facilitate packaging into chromatin structures within the nucleus. Chromosomes ensure the accurate transmission of genetic information during cell division via mitosis, which partitions duplicated chromosomes to daughter cells for growth and repair, and meiosis, which segregates homologous chromosomes to produce haploid gametes. Basic terminology describes normal and abnormal ploidy states: diploidy refers to the typical two complete sets of chromosomes in somatic cells (euploidy), aneuploidy denotes deviations such as extra or missing chromosomes leading to imbalances, and polyploidy involves more than two sets, often observed in plants but rare and pathological in humans. Cytogenetics originated in the early 20th century from the merger of cytology, the study of cell structure, and genetics, the study of heredity, particularly through observations of chromosome behavior in germ cells aligning with Mendel's laws.

Chromosomal Structure and Behavior

Chromosomes are linear structures composed primarily of DNA and histone proteins, organized into chromatin that condenses into discrete units during cell division. The basic components include the centromere, a constricted region that serves as the attachment point for spindle fibers during mitosis and meiosis; telomeres, specialized nucleotide sequences at the ends of chromosomes that protect against degradation and fusion; and the p and q arms, where the p arm denotes the shorter arm and the q arm the longer arm, separated by the centromere. Chromatin exists in two forms: euchromatin, which is loosely packed and transcriptionally active, allowing access to genetic information; and heterochromatin, which is densely packed and generally transcriptionally silent, often found in regions like centromeres and telomeres. Visualization of chromosome structure relies on techniques that highlight these components during , when chromosomes are most condensed and observable. Giemsa , a basic differential method, produces characteristic light and dark bands (G-bands) on metaphase chromosomes by preferentially staining AT-rich regions darkly and GC-rich regions lightly, enabling identification of individual based on banding patterns. This reveals the overall morphology, including arm lengths and position, which vary across the 23 pairs of human chromosomes. During mitosis, chromosomes undergo dynamic changes to ensure accurate segregation. In prophase, chromosomes condense from diffuse chromatin into compact structures facilitated by condensin proteins, which loop and fold DNA to achieve a 10,000-fold compaction. Kinetochore proteins assemble at the centromere, allowing attachment to microtubules of the mitotic spindle during prometaphase; bi-orientation ensures each sister chromatid connects to opposite spindle poles. In anaphase, cohesion between sister chromatids is cleaved by separase, enabling spindle forces to pull chromatids to opposite poles, followed by decondensation in telophase. In , exhibit behaviors that promote genetic diversity and halve the number for formation. pair during I, forming synaptonemal complexes that align non-sister chromatids for crossing over, where reciprocal exchanges of segments occur at chiasmata, facilitating recombination. This reduction division proceeds through two stages: I segregates homologous pairs to opposite poles, reducing the diploid (2n) set to haploid (n); II then separates , akin to , yielding four haploid . Chromosomal aberrations disrupt normal structure or number, potentially leading to imbalances in genetic material. Structural aberrations include deletions (loss of a segment), duplications (extra copy of a segment), inversions (reversal of a segment's ), and translocations ( between non-homologous chromosomes). Numerical aberrations involve changes in chromosome count, such as (loss of one chromosome, resulting in 2n-1) or (gain of one, resulting in 2n+1). Idiograms and chromosome maps provide standardized visual representations of these structures. An idiogram is a schematic diagram depicting chromosomes in a standardized, idealized form, ordered by size and shape with banded regions indicated, serving as a reference for locating and aberrations. Chromosome maps extend this by integrating cytogenetic bands with molecular data, such as positions, to create detailed frameworks for genomic analysis.

Historical Development

Early Observations

The foundations of cytogenetics were laid in the late through pioneering cytological observations that first identified and characterized chromosomes as distinct cellular structures. In 1879, German anatomist described thread-like bodies within the nuclei of embryo cells during division, using improved techniques to visualize their behavior in ; he termed these structures "chromatin threads," noting their longitudinal splitting and equitable distribution to daughter cells. This work built on earlier by highlighting the dynamic role of these elements in , though their hereditary significance remained unclear at the time. Nearly a decade later, in 1888, anatomist Heinrich Waldeyer-Hartz formalized the term "" (from Greek roots meaning "colored body") to denote these stained nuclear filaments observed in eukaryotic cells, emphasizing their consistent presence across dividing tissues. By the early , researchers began connecting chromosomes to , establishing cytogenetics as a bridge between cytology and . In 1902, and independently proposed the chromosome theory of , positing that chromosomes serve as physical carriers of hereditary units (later called genes), based on observations of chromosome segregation during in grasshopper spermatocytes and sea urchin embryos, respectively; this theory aligned Mendel's laws with chromosomal behavior, suggesting that parental traits are transmitted via specific chromosome pairs. Experimental confirmation came in 1910 from Thomas Hunt Morgan's studies on the Drosophila melanogaster, where he identified a white-eyed linked to sex, demonstrating that genes reside on specific chromosomes—namely, the —through breeding patterns that followed chromosomal rather than independent assortment. These findings solidified the idea that chromosomes are linear bearers of genetic information, paving the way for . Plant cytogenetics emerged concurrently, with early investigations revealing chromosomal variations influencing traits like pigmentation. Barbara McClintock's pre-1940s research at focused on (Zea mays) chromosome structure and behavior, including cytological analysis of kernel color patterns in the 1920s and 1930s; her 1931 observations of a heterochromatic knob on and studies of chromosomal aberrations in pigmentation mutants laid groundwork for understanding genetic instability, though the full mechanism of was not yet elucidated. These efforts highlighted plants as model systems for cytogenetic studies due to their visible chromosomal polymorphisms. Initial attempts to determine the human chromosome number yielded conflicting results, reflecting technical limitations in early and . In the , Theophilus Painter estimated 48 chromosomes based on testicular cell counts, a figure widely accepted but later challenged by inconsistencies in observations; alternative counts ranged around 46 to 48, with debates persisting until improved techniques in the confirmed 46 as the diploid number.

Mid-20th Century Advances

In the mid-1950s, significant progress in cytogenetics was marked by the accurate determination of the diploid chromosome number as 46, overturning the long-held belief of 48 chromosomes that had persisted since the . Joe Hin Tjio and Albert Levan achieved this breakthrough through meticulous analysis of cultured cells using improved hypotonic and colchicine-based techniques, which minimized chromosome contraction and scattering during preparation. Their findings, published in 1956, provided the foundation for modern karyotyping and revealed the 22 pairs of autosomes plus one pair of . Parallel advances in plant cytogenetics during the 1940s and 1950s were driven by Barbara McClintock's pioneering studies on , where she identified transposable elements—mobile genetic units capable of altering and structure. Through detailed cytological observations of breakage and variegated kernel phenotypes in lines, McClintock demonstrated that these "controlling elements" could insert and excise within the genome, influencing traits like pigmentation. Her work, culminating in key publications from the late 1940s to 1950s, was initially met with skepticism but later recognized as a cornerstone of genetic regulation, earning her the 1983 Nobel Prize in Physiology or Medicine. Theodosius Dobzhansky's research on in the 1930s through 1950s further solidified cytogenetics' role in by revealing chromosomal inversions as key drivers of natural variation and . Using polytene chromosomes, Dobzhansky and collaborators mapped paracentric inversions in species like , showing their prevalence in wild populations and association with ecological adaptations. These studies, including extensive surveys of inversion polymorphisms across geographic regions, underscored how structural chromosomal changes maintain without disrupting . Tjio extended his cytogenetic techniques to early mammalian studies in the , analyzing complements in cells and species like to refine preparation methods for higher eukaryotes. His work on karyotypes helped establish baseline diploid numbers and morphological details, facilitating comparative cytogenetics across mammals. These analyses built on models, where Tjio and Levan optimized pretreatments like oxyquinoline to achieve clearer spreads. By the 1970s, the advent of chromosome banding techniques revolutionized cytogenetic resolution, with emerging as a pivotal method for identifying individual . Developed through and Giemsa staining protocols, produced consistent dark and light band patterns reflecting DNA base composition differences, enabling precise homolog pairing and abnormality detection. The 1971 Conference standardized these banding patterns, establishing an international that formalized cytogenetic reporting and accelerated into chromosomal aberrations.

Core Techniques

Karyotyping Procedures

Karyotyping involves the visual examination of chromosomes at to assess their number, size, and shape, serving as a foundational in cytogenetics for detecting numerical and structural abnormalities. This process requires obtaining cells capable of division, arresting them in , preparing chromosome spreads on slides, and applying banding techniques for detailed visualization under a . The method is particularly valuable for identifying large-scale chromosomal changes, such as aneuploidies or translocations, though it has limitations in resolving smaller alterations. Cell preparation begins with collecting samples from sources rich in dividing cells, such as peripheral blood lymphocytes, bone marrow, amniotic fluid, or chorionic villi. For blood samples, approximately 0.5 mL of heparinized whole blood is inoculated into a culture medium containing phytohemagglutinin to stimulate lymphocyte proliferation, followed by incubation at 37°C for 72 hours. To arrest cells in metaphase, colchicine is added in the final hours of culture, disrupting microtubule formation and halting division at this stage where chromosomes are maximally condensed and visible. Cultures from prenatal samples, like amniotic fluid, may require longer incubation periods of 7 to 14 days to ensure sufficient cell growth. Following harvest, cells undergo hypotonic treatment with a 0.075 M (KCl) solution for 10-20 minutes to swell them and spread chromosomes. The swollen cells are then fixed multiple times (typically three) in a 3:1 :acetic acid solution to preserve structure and remove . Fixed cell suspensions are dropped onto clean slides to create spreads, allowing chromosomes to air-dry in a dispersed manner suitable for microscopic analysis. Staining enhances visibility and enables banding patterns for identification. The standard technique, using , involves treating slides with to partially digest chromosome proteins, followed by incubation in 2x and with diluted Giemsa solution, producing characteristic light and dark bands that reflect Giemsa-positive and Giemsa-negative regions. Analysis entails microscopic examination, where technicians manually count and arrange chromosomes into a karyogram, pairing homologs and ordering them by size and position. Automated systems can assist in capturing images and generating karyograms, but human verification remains essential for accuracy. Karyotyping can detect chromosomal alterations larger than 5-10 , such as deletions or duplications of that scale, but misses smaller variants below this resolution threshold. Abnormalities are described using the International System for Human Cytogenomic Nomenclature (ISCN), a standardized format that specifies chromosome number, , and structural details (e.g., 47,XX,+21 for trisomy 21). The 2024 edition of ISCN incorporates updates for integrating cytogenomic data from karyotyping with other methods, ensuring consistent reporting across laboratories.

Fluorescence In Situ Hybridization (FISH)

Fluorescence in situ hybridization (FISH) is a molecular cytogenetic technique that employs fluorescently labeled DNA or RNA probes to bind specifically to complementary target sequences on chromosomes or in interphase nuclei, enabling their visualization through fluorescence microscopy. The method exploits the principle of nucleic acid hybridization, where probes anneal to denatured chromosomal DNA under controlled conditions, allowing precise localization of genes or genomic regions within the cellular context. Originating from earlier radioactively labeled in situ hybridization techniques introduced by Pardue and Gall in 1969, FISH advanced significantly in the 1980s with the development of fluorescent labeling, as demonstrated by Pinkel et al. in 1986, which enabled quantitative detection of chromosomal abnormalities with high sensitivity. The procedure for FISH begins with the fixation and preparation of cells or tissues on slides, often utilizing spreads or nuclei from samples like , , or tumor biopsies, which may involve brief reference to standard fixation methods for chromosome immobilization. Probes are designed as cloned DNA fragments, such as bacterial artificial chromosomes () spanning 100-200 kb or synthetic , labeled directly with fluorochromes like fluorescein or indirectly via haptens such as or digoxigenin for signal amplification. The target DNA and probe are denatured simultaneously using heat (typically 70-80°C) or chemical agents like to separate strands, followed by hybridization in a humid chamber for 6-48 hours at 37°C to allow specific binding. Unbound probes are then removed through stringent washing steps with saline-sodium citrate buffers, and signals are detected either directly via the or indirectly through secondary antibodies conjugated to fluorochromes. FISH encompasses several types tailored to specific cytogenetic needs, including locus-specific probes that target individual genes or small regions (1-1000 kb) for detecting microdeletions or amplifications, and repetitive sequence probes such as those for centromeres or telomeres to assess . Whole-chromosome painting probes, derived from flow-sorted chromosomes or degenerate primers, label entire s in one color, facilitating the of structural rearrangements like translocations. Advanced multicolor variants, such as spectral karyotyping () introduced in the 1990s, use combinatorial labeling with up to 24 fluorochromes to paint each chromosome pair in a unique color spectrum, while multiplex FISH (M-FISH) employs five dyes for analysis, enabling simultaneous detection of multiple aberrations in a single hybridization. Analysis of FISH results relies on epifluorescence or equipped with filter sets matching the emission spectra of the fluorochromes, where bound probes appear as discrete fluorescent spots or domains corresponding to the target loci on chromosomes or in nuclei. Quantitative assessment involves counting signal numbers and positions to identify numerical changes (e.g., ) or structural variants (e.g., fusions indicating translocations), with software aiding in image capture and for multicolor setups. The technique achieves a of approximately 100 in cells and 1-5 Mb on spreads, allowing detection of submicroscopic alterations not visible by conventional banding. Compared to traditional karyotyping, provides superior detection of submicroscopic chromosomal changes, such as microdeletions or cryptic translocations, and permits analysis of non-dividing cells without the need for prolonged , yielding results in 1-3 days. It enhances by integrating molecular specificity with cytological context, making it indispensable for precise diagnostics in genetic disorders and .

Advanced Methods

Molecular Cytogenetic Tools

Molecular cytogenetic tools represent an evolution of traditional (FISH) techniques, integrating with to enable genome-wide analysis of chromosomal alterations at higher resolution. These methods, developed primarily in the and 2000s, allow for the detection of copy number variations (CNVs) and structural rearrangements without relying solely on spreads, facilitating applications in and constitutional . By combining DNA labeling, hybridization, and advanced imaging, they provide insights into genomic imbalances that complement classical cytogenetic approaches. Comparative genomic hybridization (CGH), introduced in the early 1990s, is a technique that compares the DNA content of a test sample (e.g., from a tumor) to a reference genome by co-hybridizing differentially labeled DNAs onto normal metaphase chromosomes. This method generates a ratio profile along the chromosomes, highlighting regions of gain or loss that indicate CNVs, such as amplifications or deletions, with a resolution of approximately 5-10 Mb. CGH has been instrumental in mapping recurrent chromosomal changes in solid tumors, bypassing the need to culture tumor cells directly. Array comparative genomic hybridization (array CGH), an advancement from the 2000s, replaces chromosomes with a of genomic clones or , enabling higher-resolution detection of CNVs down to about 1 in modern iterations. In this approach, test and reference DNAs are labeled with different fluorophores, hybridized to the array, and analyzed for intensity ratios to identify imbalances; notably, it requires only extracted DNA, eliminating the need for or viable s. This tool has revolutionized prenatal diagnostics and by allowing rapid, of genomic copy number alterations. Spectral karyotyping (SKY), developed in the mid-1990s, employs a combinatorial labeling strategy with multiple fluorophores to "paint" each in a unique , visualized through Fourier spectroscopy and imaging software. This 24-color technique for human chromosomes facilitates the identification of complex chromosomal rearrangements, marker chromosomes, and homogeneously staining regions in spreads, particularly useful in analyzing tumor karyotypes with multiple aberrations. SKY builds on by providing a whole-genome overview, distinguishing chromosomes and their derivatives even in highly rearranged genomes. Fiber-FISH extends to linearly extended chromatin fibers or DNA molecules, achieving resolutions of 1-5 kb for precise mapping of repetitive sequences, large inserts, or clone contigs. By stretching DNA on a slide and hybridizing locus-specific probes, this method visualizes the physical order and spacing of genomic elements along extended fibers, aiding in the assembly of large-scale maps and detection of duplications or deletions in extended regions. It is particularly valuable for validating sequencing data or studying highly repetitive genomic areas like telomeres. Despite their advancements, molecular cytogenetic tools like CGH and array CGH have limitations, including an inability to detect balanced translocations or inversions that do not alter DNA copy number, necessitating complementary methods such as karyotyping or sequencing for comprehensive analysis. Additionally, resolution constraints in traditional CGH and potential artifacts from probe cross-hybridization can complicate interpretation in heterogeneous samples.

High-Throughput Approaches

High-throughput approaches in cytogenetics leverage advanced sequencing technologies and computational tools to analyze chromosomal structures at scale, enabling the detection of structural variants (SVs) with unprecedented resolution and throughput. Optical genome mapping (OGM), developed in the 2010s, utilizes nanochannel arrays to linearize and image long DNA molecules labeled at specific sequence motifs, providing long-range chromosomal scaffolding without the need for assembly. This technique excels at identifying SVs larger than 500 base pairs, including balanced translocations and inversions that are challenging for traditional methods, by generating genome-wide maps with kilobase to megabase resolution. In constitutional and somatic applications, OGM has demonstrated superior sensitivity for complex rearrangements, often complementing karyotyping by revealing cryptic abnormalities in up to 58% of cases where standard techniques fall short. Next-generation sequencing (NGS) has transformed cytogenetic analysis through methods like mate-pair sequencing, which generates long-insert paired-end reads to detect SVs by identifying discordant read pairs and split reads across breakpoints. Mate-pair libraries, typically with inserts of 2-5 kb, facilitate the precise mapping of chromosomal aberrations, achieving over 90% detection of cytogenetically visible breakpoints and refining their locations to sub-band resolution. Integration of NGS with traditional karyotyping enhances validation, as mate-pair data can confirm and delineate variants observed microscopically, improving diagnostic accuracy in scenarios like where SVs drive oncogenesis. Single-cell cytogenomics extends these capabilities to heterogeneous samples, employing single-cell whole-genome sequencing (scWGS) or single-cell sequencing (scRNA-seq) to profile and chromosomal imbalances at the individual cell level. scWGS provides genome-wide copy number profiles, detecting in preimplantation embryos with high fidelity, while scRNA-seq infers chromosomal arm-level gains and losses from expression imbalances across hundreds of genes per . In tumor contexts, these approaches reveal intratumor heterogeneity, identifying subpopulations that correlate with therapeutic , as validated in studies of hematopoietic malignancies. Bioinformatics pipelines are essential for processing high-throughput cytogenetic data, with tools like DELLY and enabling robust SV calling from NGS reads by integrating split-read and paired-end evidence. DELLY employs a Poisson-based model to cluster anomalous reads for deletions, insertions, and inversions, while uses active conditioning to rapidly detect medium-sized indels and large s in and samples. These algorithms achieve single-nucleotide but prioritize cytogenetic-scale events, such as translocations spanning kilobases, with consensus approaches combining multiple callers yielding precision above 80% in low-coverage whole-genome sequencing. In the 2020s, (AI) has advanced automated karyotyping by employing models to segment and classify chromosomes from microscopic images, reducing manual analysis time from hours to minutes. AI-guided systems, such as those supporting G-, Q-, and R-banding across diverse sample types, improve efficiency at scale in clinical labs, with studies reporting enhanced accuracy in abnormality detection and standardization of interpretations. For instance, convolutional neural networks in tools like KaryoXpert achieve instance-level chromosome classification with over 95% accuracy, facilitating in research cohorts.

Applications in Research

Plant and Animal Studies

Cytogenetics has played a pivotal role in understanding in , particularly in crop species like (Triticum aestivum), which possesses 42 chromosomes resulting from successive hybridization events and duplication. The hexaploid of bread arose through allopolyploidization involving ancestors from the genera Aegilops and Triticum, where the A genome (2n=14) originated from Triticum urartu, the B genome (2n=28) from an unidentified Aegilops species, and the D genome (2n=14) from Aegilops tauschii. This polyploid structure enhances and adaptability, enabling wheat's and yield improvements, as demonstrated by cytogenetic analyses of chromosome pairing during that reveal homeologous relationships among subgenomes. In maize (Zea mays), cytogenetic studies by Barbara McClintock in the mid-20th century uncovered transposable elements, or "jumping genes," through observations of chromosome breakage and variegated kernel phenotypes. McClintock identified the Activator-Dissociation (Ac-Ds) system, where Ds elements insert into genes like those controlling pigment production, causing mutable alleles that revert under Ac influence, thus providing a mechanism for gene tagging and epigenetic regulation. These findings, derived from detailed karyotypic and phenotypic mapping, revolutionized understanding of genome dynamics and transposon-mediated evolution in plants. Animal cytogenetics has advanced through model organisms like Drosophila pseudoobscura, where documented paracentric inversions in natural populations during the 1930s and 1970s, revealing their role in suppressing recombination and maintaining adaptive complexes. In salivary gland polytene chromosomes, these inversions formed clinal distributions across geographic populations, linking chromosomal polymorphisms to evolutionary adaptation and . In mice (Mus musculus), karyotyping has been essential for studies, identifying induced chromosomal aberrations such as translocations and deletions following exposure to or chemicals, which map hotspots and inform function in mammalian development. Cytogenetic research has illuminated chromosome evolution in animals, exemplified by telomeric fusions that reduced the primate karyotype from 48 to 46 chromosomes in humans via the head-to-head fusion of two ancestral acrocentric chromosomes now forming . Remnants of telomeres and a vestigial at 2q13-2q14.1 confirm this event, distinguishing human cytogenetics from great apes like chimpanzees. Sex chromosome heterogamety further highlights evolutionary divergence, with XY systems in mammals—where males are heterogametic () and females homogametic ()—contrasting ZW systems in , where females are heterogametic (ZW) and males homogametic (ZZ); these arose independently in amniotes, with conserved content on Z/W and X/Y despite distinct origins. In agricultural breeding, (FISH) facilitates the introgression of alien genes from wild relatives into crops, enabling precise monitoring of segments for traits like resistance. For instance, FISH probes target rye (Secale cereale) transferred to , allowing visualization of translocation breakpoints and stable integration of beneficial loci while minimizing linkage drag, thus enhancing crop resilience without disrupting native genomes.

Population Genetics

Cytogenetics plays a crucial role in by revealing structural variations in chromosomes that influence , , and evolutionary processes in natural populations. Chromosomal polymorphisms, such as inversions and translocations, serve as visible markers of that can be directly observed through techniques like karyotyping, allowing researchers to track frequencies and across populations. These variants often suppress recombination, preserving co-adapted gene complexes and contributing to local in heterogeneous environments. In species, chromosomal inversions exemplify how polymorphisms drive , with over 70 inversions documented in Drosophila subobscura alone, many associated with clinal variation in traits like cold tolerance and migration behavior. For instance, inversions on the third chromosome in , such as In(3R)Payne, have been linked to latitudinal clines in fitness-related traits, maintaining through recombination suppression and facilitating responses to environmental gradients. These polymorphisms, first noted in mid-20th century studies, highlight the long-term stability of inversion frequencies in natural populations under selective pressures. Karyotypic variation, including changes in chromosome number and structure, further informs speciation processes, as seen in grasshoppers where clinal patterns of fusion and fission events correlate with geographic isolation. In the Wood White butterfly (Leptidea sinapis), chromosome numbers vary clinally from 2n=106 in to 2n=56 in eastern across a 6000 km range, with these polymorphisms reducing hybrid fertility and promoting between populations. Such variations underscore the role of cytogenetic rearrangements in generating barriers to , accelerating divergence in natural settings. Cytogenetic mechanisms also underlie phenomena like and segregation distortion, where selfish genetic elements bias transmission by disrupting normal segregation during . In , the Segregation Distorter (SD) complex on uses inversions to suppress recombination, protecting linked distorter alleles that cause dysfunction in sensitive , thereby elevating transmission rates up to 99% in heterozygous males. This distortion maintains polymorphisms in populations but can lead to evolutionary arms races with suppressors, influencing overall . In conservation genetics, cytogenetic monitoring assesses chromosomal integrity in , particularly through chromosome counts and polymorphism detection to identify hybridization risks or inbreeding effects. Felids, such as and , exhibit a highly conserved with 2n=38 across genera, enabling straightforward comparisons to detect anomalies in captive or wild populations and guide breeding programs for genetic health. Quantitative insights from cytogenetic data include estimates of heterozygosity rates for structural variants, often exceeding 20-30% in polymorphic populations like , and elevated (LD) within rearranged regions due to recombination suppression. For example, inversion heterozygotes show LD decay rates 10-100 times slower than collinear segments, allowing inference of historical recombination and population from surveys. These metrics provide scalable measures of without sequencing, essential for tracking evolutionary dynamics.

Medical and Clinical Uses

Detection of Chromosomal Abnormalities

Cytogenetic techniques play a crucial role in diagnosing chromosomal abnormalities associated with congenital and developmental disorders, enabling early identification of genetic risks in human pregnancies and affected individuals. These methods primarily involve analyzing structure and number to detect aneuploidies, deletions, duplications, and other structural variants that can lead to syndromes impacting , , and overall . By providing direct of chromosomes, cytogenetics offers a foundational approach to clinical , often integrated with molecular tools for enhanced precision. Prenatal testing for chromosomal abnormalities typically employs invasive procedures such as and (CVS), which allow for the collection of fetal cells suitable for cytogenetic analysis. , performed between 15 and 20 weeks of , involves extracting containing fetal cells, which are then cultured and subjected to karyotyping to reveal numerical abnormalities like 21, responsible for . Similarly, CVS, conducted earlier at 10 to 13 weeks, samples placental tissue for the same analyses, detecting conditions such as (Edwards syndrome) and 13 () with high accuracy. Chromosomal microarray analysis () is recommended, particularly for fetuses with structural abnormalities or when a normal karyotype is found, to identify submicroscopic copy number variations with higher resolution than karyotyping alone. () complements karyotyping in these samples by using fluorescent probes to target specific chromosomal regions, enabling rapid detection of aneuploidies within 24-48 hours, which is critical for timely clinical decisions. These techniques have detection rates exceeding 99% for major trisomies when combined, though they carry a small risk of procedure-related (approximately 0.1-0.5%). Postnatal diagnosis of chromosomal abnormalities often relies on peripheral blood karyotyping, where lymphocytes are stimulated, cultured, and analyzed to identify sex chromosome disorders and other aneuploidies; CMA may also be used for higher-resolution detection of copy number variants. For instance, Turner syndrome, characterized by a 45,X karyotype, is diagnosed through this method in females presenting with short stature, ovarian dysgenesis, and cardiac anomalies, confirming the monosomy X in about 50-60% of cases while mosaicism accounts for the rest. In males, Klinefelter syndrome (47,XXY) is detected via blood karyotypes, revealing the extra X chromosome that leads to hypogonadism, infertility, and increased risk of metabolic issues; this approach identifies over 90% of cases when clinically suspected. These analyses typically take 7-14 days but provide comprehensive chromosome profiles, guiding management strategies like hormone replacement therapy. For microdeletion syndromes, FISH serves as a targeted diagnostic tool to identify submicroscopic deletions not visible on standard karyotypes, though CMA offers genome-wide detection. , resulting from a 22q11.2 deletion, is diagnosed using FISH probes specific to the region, detecting the abnormality in approximately 90% of cases and linking it to features like conotruncal heart defects, , and immune deficiency. Likewise, , caused by a 7q11.23 deletion, is confirmed via FISH, which highlights the gene locus deletion responsible for supravalvular , hypercalcemia, and distinctive facial features; this method's sensitivity reaches 95-99% for these contiguous gene syndromes. Such applications underscore FISH's value in clinical settings for syndromes affecting 1 in 4,000 to 1 in 20,000 births. Non-invasive prenatal testing (NIPT) using (cfDNA) from maternal blood has revolutionized screening for chromosomal abnormalities, often serving as a first-line before confirmatory cytogenetic testing. Introduced in the early , NIPT analyzes fetal DNA fractions (typically 4-10% of total cfDNA) to detect trisomies 21, 18, and 13 with sensitivities above 99% and specificities over 99.9%, particularly in high-risk pregnancies. When positive, it prompts invasive cytogenetic follow-up like karyotyping or to rule out confined placental mosaicism or maternal abnormalities. This integration reduces the need for invasive procedures by up to 50% in screened populations while maintaining diagnostic accuracy for fetal aneuploidies. Ethical considerations in cytogenetic detection of chromosomal abnormalities emphasize , , and psychosocial support, given the profound implications of findings on and . Counseling sessions, recommended by professional guidelines, address the uncertainty of results (present in 1-2% of diagnoses), the option for pregnancy termination in prenatal cases, and the lifelong management of diagnosed syndromes. For example, discovering a trisomy 21 prenatally requires discussing developmental outcomes, with uptake rates for termination varying by region (50-90% in some studies), highlighting the need for non-directive, culturally sensitive guidance to mitigate anxiety and ensure autonomous decision-making.

Cancer Cytogenetics

Cancer cytogenetics plays a pivotal role in by identifying somatic chromosomal alterations that drive tumorigenesis and influence clinical outcomes. These changes, including translocations, amplifications, and aneuploidies, are detected through techniques such as karyotyping and (FISH), enabling the classification of tumors and guidance of therapeutic decisions. In hematologic malignancies like leukemias, specific recurrent abnormalities provide diagnostic hallmarks and prognostic insights, while in solid tumors, they reveal mechanisms of activation and genomic instability. In leukemias, the Philadelphia chromosome, resulting from the t(9;22)(q34;q11) translocation, is a defining feature of chronic myeloid leukemia (CML), observed in approximately 90-95% of cases via G-banded karyotyping. This translocation fuses the BCR gene on chromosome 22 with the ABL1 gene on chromosome 9, producing the BCR-ABL fusion oncoprotein that drives leukemogenesis. FISH enhances detection of this fusion, including variant translocations, and is particularly useful for monitoring minimal residual disease. Solid tumors exhibit diverse cytogenetic alterations, such as gene amplifications and . For instance, amplification of the HER2 (ERBB2) gene on 17q12 occurs in about 15-20% of cancers and is associated with aggressive and poorer survival, serving as a key target for therapies like . FISH is the gold standard for confirming HER2 amplification when is equivocal, following ASCO/ guidelines that define amplification as a HER2/CEP17 ratio ≥2.0. In , is prevalent in up to 70% of cases, often linked to chromosomal instability pathways that promote tumor progression and . Prognostic markers derived from cytogenetic profiles are crucial for risk stratification. In acute lymphoblastic leukemia (ALL), high hyperdiploidy (51-65 chromosomes) is a favorable feature in pediatric B-cell precursor cases, conferring improved event-free survival rates exceeding 90% due to enhanced sensitivity to antimetabolites. Conversely, complex karyotypes—defined as three or more unrelated chromosomal abnormalities—are associated with poor outcomes across various cancers, including , where they predict inferior overall survival independent of other factors. Clonal evolution in tumors can be tracked through serial cytogenetic profiling, revealing the emergence of subclones with additional abnormalities that drive progression from indolence to aggressiveness. For example, in , quantitative multicolour demonstrates phylogenetic relationships among clones, linking cytogenetic changes to relapse and resistance. Cytogenetic findings directly inform targeted therapies, particularly tyrosine kinase inhibitors (TKIs) for translocation-driven cancers. In CML, the guides , which inhibits the oncogenic kinase and achieves complete cytogenetic responses in over 80% of chronic-phase patients, transforming CML into a manageable . Similar principles apply to other translocations, such as PML-RARA in , where all-trans targets the fusion product identified cytogenetically.

Integration with Genomics

The integration of cytogenetics with genomics has revolutionized the analysis of chromosomal abnormalities by combining traditional karyotyping and fluorescence in situ hybridization (FISH) with next-generation sequencing (NGS) technologies, enabling hybrid approaches that validate cytogenetic findings at the molecular level. Short-read NGS confirms structural variants (SVs) identified via classical methods, while long-read sequencing resolves complex rearrangements that karyotyping alone cannot detect, such as insertions or inversions smaller than 5 Mb. For instance, in clinical diagnostics, NGS validates FISH-detected fusions like BCR::ABL1 in chronic myeloid leukemia, providing precise breakpoint mapping to guide targeted therapies. A key advantage of these hybrid methods is their ability to uncover hidden complexities in apparently balanced translocations, which traditional cytogenetics often misses due to resolution limits. Whole-genome sequencing (WGS) applied to carriers of balanced rearrangements reveals that approximately 37% harbor cryptic imbalances or gene disruptions not visible by karyotyping, such as small deletions at breakpoints that contribute to phenotypic abnormalities. Long-read NGS, including PacBio's , excels at phasing these SVs across haplotypes, enabling accurate reconstruction of translocation events and their functional impacts in the 2020s. This has improved prenatal and postnatal diagnostics by identifying pathogenic variants in up to 58% more cases when integrated with optical genome mapping. Epigenetic cytogenetics further bridges these fields by incorporating ChIP-seq to map structure and modifications alongside chromosomal analyses, revealing how marks and influence architecture. ChIP-seq, which immunoprecipitates -bound proteins followed by sequencing, identifies genome-wide binding sites of epigenetic regulators, linking them to large-scale chromosomal domains like regions observed in cytogenetic banding. When combined with cytogenetic tools such as for 3D conformation, this approach elucidates interactions between distant chromosomal loci, as demonstrated in studies of enhancer-promoter looping in cancer cells. Such integrations provide insights into how epigenetic alterations drive chromosomal instability without altering DNA sequence. In , cytogenomic profiling—merging cytogenetic SV detection with genomic sequencing—supports precision through multidisciplinary tumor boards that interpret multi-omics data for tailored treatments. In the , these boards use comprehensive genomic profiling to match patients with therapies targeting specific fusions or copy number variations. For example, integrating karyotype-derived SVs with NGS identifies resistance mechanisms in , guiding inhibitors like for BCR::ABL1-positive cases. Managing the big data generated by these integrations poses challenges, addressed by databases like COSMIC, which curates over 10,000 whole-genome sequenced samples to catalog cytogenetic variants including translocations and inversions. COSMIC's structural variant signatures, derived from validated calls across 16 tissue types, facilitate querying of prevalence and mutational processes, though restricted access to clinical metadata limits full integration. This resource enables researchers to correlate cytogenetic findings with genomic contexts, overcoming silos in for better variant interpretation in .

Future Challenges and Innovations

One major challenge in cytogenetics is the declining reliance on classical karyotyping, as next-generation sequencing (NGS) technologies offer higher resolution for detecting chromosomal abnormalities, prompting questions about the continued development of traditional cytogenetic methods. This shift has reduced the use of karyotyping in routine diagnostics, particularly for conditions like , where NGS provides rapid genomic profiling with superior . Concurrently, detecting structural variants (SVs) remains problematic due to their and the limitations of current analytical tools, which struggle with long SVs exceeding read lengths and intratumor heterogeneity in cancer samples. Efforts to address this include low-pass whole-genome sequencing, which enables cost-effective SV detection at reduced sequencing depths while maintaining reliability for variants. Innovations in CRISPR-based approaches are expanding cytogenetic capabilities by enabling precise and live visualization of chromosomal structures. CRISPR-Cas systems, particularly when engineered for , target repetitive genomic regions such as centromeres and telomeres to track three-dimensional dynamics in , offering insights into organization beyond static karyotypes. Complementing this, (AI) is transforming cytogenetic analysis through automated karyotyping, where algorithms classify chromosomes with high accuracy, reducing manual effort and enabling real-time processing during procedures like biopsies. In clinical settings, cytogenomics is poised to become routine in in vitro fertilization (IVF) for preimplantation (PGT), where NGS-based chromosomal screening identifies aneuploidies in embryos to enhance implantation success and reduce risks. Similarly, integrating chromosomal variants into could personalize drug responses by accounting for structural changes like translocations that influence medication efficacy, as seen in guidelines incorporating such variants for precision therapy. Ethical concerns loom large with the rise of (DTC) cytogenetic testing, which risks misinterpretation of results due to limited clinical utility and high false-positive rates for variants classified as high-risk. privacy in genomic databases exacerbates these issues, as re-identification attacks on seemingly datasets could expose sensitive chromosomal information, necessitating robust frameworks for shared cytogenetic . Looking ahead, quantum computing holds promise for modeling complex chromosome structures, such as chromatin folding, by simulating three-dimensional genome interactions that classical computers struggle to compute efficiently. In space biology, cytogenetics faces unique challenges from radiation-induced chromosomal aberrations, where ionizing particles cause DNA damage and translocations, informing protective strategies for long-duration missions.

References

  1. [1]
    Cytogenetics - National Human Genome Research Institute
    Cytogenetics is a branch of biology focused on the study of chromosomes and their inheritance, especially as applied to medical genetics.
  2. [2]
    Genetics, Cytogenetic Testing and Conventional Karyotype - NCBI
    Aug 8, 2023 · Cytogenetic testing is the examination of chromosomes to determine chromosome abnormalities such as aneuploidy and structural abnormalities.
  3. [3]
    Cytogenetics: Past, Present And Future - PMC - PubMed Central - NIH
    Cytogenetics is the study of the structure and properties of chromosomes, their behaviour during somatic cell division during growth and development (mitosis),
  4. [4]
    History and evolution of cytogenetics - PMC - PubMed Central - NIH
    Mar 20, 2015 · The history of human cytogenetics has been punctuated by the introduction of new technology which on each occasion has led to the discovery of ...Introduction · Chromosome Structure · The Emergence Of Human...
  5. [5]
    Cytogenetic Techniques - Hsieh - Wiley Online Library
    Mar 15, 2011 · Cytogenetics is the study of the genetic material in cells at the light microscopic level, with the goal of achieving optimal resolution for ...
  6. [6]
    Definition of cytogenetics - NCI Dictionary of Cancer Terms
    Cytogenetics involves testing samples of tissue, blood, or bone marrow in a laboratory to look for changes in chromosomes.
  7. [7]
    Cytogenetics and Cytogenomics in Clinical Diagnostics: Genome ...
    This review addresses the central question of how nuclear genome architecture contributes to disease mechanisms and diagnostics, and how technological advances ...
  8. [8]
    Cytogenetics - an overview | ScienceDirect Topics
    Cytogenetics is the study of chromosome structure and number, and the diseases caused by their abnormalities, including changes in structure or number.Missing: scope | Show results with:scope
  9. [9]
    Cytogenetics Testing - ARUP Laboratories
    Cytogenetics testing aids in the diagnosis of heritable genetic abnormalities and is also used to detect acquired or somatic abnormalities relevant for the ...Why Choose Arup? · Genetic Counselor Services · Support Of Optimal Patient...
  10. [10]
    Overview of Clinical Cytogenetics - 2016 - Wiley Online Library
    Apr 1, 2016 · Chromosome analysis can be performed prenatally to provide information about the developing fetus or postnatally to aid in diagnosis of ...<|control11|><|separator|>
  11. [11]
    chromatin | Learn Science at Scitable - Nature
    Chromatin is a complex of DNA and proteins that forms chromosomes within the nucleus of eukaryotic cells. Nuclear DNA does not appear in free linear strands ...<|control11|><|separator|>
  12. [12]
    Chromosome Dynamics during Mitosis - PMC - NIH
    The primary goal of mitosis is to partition duplicated chromosomes into daughter cells. Eukaryotic chromosomes are equipped with two distinct classes of ...
  13. [13]
    Re-starting life: Fertilization and the transition from meiosis to mitosis
    During meiosis I, pairs of homologous chromosomes are segregated so that each daughter cell contains half the number of chromosomes as the parent.Fertilization · Sperm-Egg Binding · Chromatin Remodelling<|control11|><|separator|>
  14. [14]
    Preimplantation Genetic Testing for Chromosomal Abnormalities - NIH
    Diploid cells normally contain 46 chromosomes, a state known as euploidy. Aneuploidy is an altered condition involving a deviation in copy number from multiples ...
  15. [15]
    Polyploidy | Learn Science at Scitable - Nature
    Polyploidy is the heritable condition of possessing more than two complete sets of chromosomes. Polyploids are common among plants, as well as among certain ...Missing: terminology | Show results with:terminology
  16. [16]
    Genetics, Chromosomes - StatPearls - NCBI Bookshelf - NIH
    A chromosome is a DNA molecule that contains the genetic information for an organism. The chromosomal structure is composed of the organism's DNA and special ...
  17. [17]
    How do geneticists indicate the location of a gene? - MedlinePlus
    Mar 26, 2021 · By convention, the shorter arm is called p, and the longer arm is called q. The chromosome arm is the second part of the gene's address.
  18. [18]
    The Global Structure of Chromosomes - Molecular Biology of the Cell
    ... telomeres have additional structural features that distinguish them from other parts of chromosomes. Centromeres Are Also Packaged into Heterochromatin.
  19. [19]
    1 - Chromosome Structure - EdTech Books
    The centromere and telomere regions of chromosomes are composed of heterochromatin. Euchromatin. Euchromatin refers to the loosely condensed regions along the ...
  20. [20]
  21. [21]
    Chromosome Preparation From Cultured Cells - PMC - NIH
    Jan 28, 2014 · Chromosomes are analyzed at the metaphase stage of mitosis, when ... staining with Giemsa to create characteristic light and dark bands.
  22. [22]
  23. [23]
    Mechanisms of chromosome behaviour during mitosis - PMC
    Proper chromosome segregation requires that both kinetochores on each chromosome attach to spindle micro tubules. Furthermore, chromosomes must be bi ...
  24. [24]
  25. [25]
    Meiosis - Molecular Biology of the Cell - NCBI Bookshelf - NIH
    During chromosomal crossing-over, the DNA double helix is broken in both a maternal chromatid and a homologous paternal chromatid, so as to exchange fragments ...
  26. [26]
    Genetics, Meiosis - StatPearls - NCBI Bookshelf - NIH
    When homologous chromosomes come together to form tetrads, the arms of the chromatids can swap at random, creating many more possibilities for genetic variation ...
  27. [27]
    Chromosome Abnormalities Fact Sheet
    Aug 15, 2020 · Deletions: A portion of the chromosome is missing or deleted. · Duplications: A portion of the chromosome is duplicated, resulting in extra ...
  28. [28]
    CHROMOSOMAL ABNORMALITIES - Understanding Genetics - NCBI
    Unbalanced rearrangements include deletions, duplications, or insertions of a chromosomal segment. Ring chromosomes can result when a chromosome undergoes two ...
  29. [29]
    Genetics, Chromosome Abnormalities - StatPearls - NCBI Bookshelf
    Apr 24, 2023 · A chromosomal abnormality, or chromosomal aberration, is a disorder characterized by a morphological or numerical alteration in single or multiple chromosomes.Missing: terminology diploidy
  30. [30]
  31. [31]
    Chromosomes and Cytogenetics | Learn Science at Scitable - Nature
    Cytogeneticists can then use coordinates on these rough chromosome maps, or idiograms, to identify the positions of structural abnormalities, including ...
  32. [32]
    1879: Mitosis observed - National Human Genome Research Institute
    Apr 22, 2013 · Flemming observed cell division in salamander embryos, where cells divide at fixed intervals. He developed a way to stain chromosomes to observe ...
  33. [33]
    1902: Chromosome Theory of Heredity
    Apr 22, 2013 · Sutton had been observing grasshopper cells, where chromosomes have quite distinct shapes. He published his findings in 1902 and a year later, ...
  34. [34]
    “Sex Limited Inheritance in Drosophila” (1910), by Thomas Hunt ...
    May 22, 2017 · That result indicated that eye color and sex were both tied to chromosomes and helped Morgan and colleagues establish that chromosomes carry the ...
  35. [35]
    THE CHROMOSOME NUMBER OF MAN - TJIO - 1956 - Hereditas
    Levan, A. 1956. Chromosome studies on some human tumors and tissues of normal origin, grown in vivo and in vitro at the Sloan-Kettering Institute.
  36. [36]
    Joe Hin Tjio Counts Chromosomes - Science History Institute
    Jun 6, 2024 · Humans don't have 48 chromosomes. They have 46. Tjio had had a solid but not spectacular career before this, mostly working under Levan's ...Missing: estimates | Show results with:estimates
  37. [37]
  38. [38]
    Karyotype — Knowledge Hub - Genomics Education Programme
    Cell culturing is required for karyotyping. This can take anything from three days (blood and bone marrow) up to 7 to 14 days (skin and prenatal samples). The ...
  39. [39]
    Preparation Of Peripheral Blood Cells For Chromosome Analysis
    Test procedure · Inoculate approximately 0.5 mL of heparinized whole blood into a glass or plastic tube with 10 mL of medium. · Incubate the culture at 37°C in 5% ...
  40. [40]
    Protocol for preparation and staining of chromosomes isolated from ...
    Feb 22, 2024 · Table 1 shows a comprehensive overview of conventional and molecular cytogenetics with reference to the corresponding steps in the protocol.
  41. [41]
    ISCN – Interpretation & communication of human cytogenomic ...
    A new nomenclature is introduced specifically for genomic mapping in the diagnostic setting taking elements of karyotyping, microarray and region-specific ...ISCN 2020 (PDF) · ISCN 2024 · ISCN 2016 (PDF) · ISCN 2009 (PDF)
  42. [42]
    ISCN 2024: An International System for Human Cytogenomic ...
    $$120.00ISCN 2024 is an indispensable reference volume for human cytogeneticists, molecular geneticists, clinical geneticists, technicians, and students.
  43. [43]
  44. [44]
    Technical Review: In Situ Hybridization - Jensen - 2014
    May 9, 2014 · Fluorescence In Situ Hybridization​​ FISH is an effective technique that enables direct visualization of genetic alterations in the cell.Abstract · Background · Multicolor Fish
  45. [45]
    Fluorescence In Situ Hybridization (FISH) and Its Applications - PMC
    Feb 10, 2017 · FISH is the most convincing technique for locating the specific DNA sequences, diagnosis of genetic diseases, gene mapping, and identification of novel ...
  46. [46]
    Fluorescent in situ hybridisation (FISH) — Knowledge Hub
    FISH is a cytogenetic technique using DNA probes to detect specific sequences on chromosomes, providing information on copy number and position.
  47. [47]
    Comparative Genomic Hybridization for Molecular Cytogenetic ...
    Abstract. Comparative genomic hybridization produces a map of DNA sequence copy number as a function of chromosomal location throughout the entire genome.
  48. [48]
    Multicolor Spectral Karyotyping of Human Chromosomes - Science
    The simultaneous and unequivocal discernment of all human chromosomes in different colors would be of significant clinical and biologic importance.Missing: original | Show results with:original
  49. [49]
    Comparative genomic hybridization: Uses and limitations
    Based on its technical limitations, there is little reason to apply CGH to chromosomes of metaphase cells in routine diagnostic settings.
  50. [50]
    Optical Genome Mapping as a Next-Generation Cytogenomic Tool ...
    Next generation cytogenetics: Genome-imaging enables comprehensive structural variant detection for 100 constitutional chromosomal aberrations in 85 samples.Missing: paper | Show results with:paper
  51. [51]
    Optical genome mapping refines cytogenetic diagnostics, prognostic ...
    Sep 2, 2022 · Optical genome mapping refines cytogenetic diagnostics, prognostic stratification and provides new molecular insights in adult MDS/AML patients.Missing: paper | Show results with:paper
  52. [52]
    Clinical Utility of Optical Genome Mapping as an Additional Tool in a ...
    Apr 25, 2025 · OGM identified additional cytogenomic abnormalities in 58% of cases, with 15% of cases showing findings that impacted diagnosis, prognosis, or treatment ...Missing: paper | Show results with:paper
  53. [53]
    Mate pair sequencing for the detection of chromosomal aberrations ...
    Oct 9, 2013 · We demonstrate that mate pair sequencing enables the rapid identification and delineation of structural variants and has added value for the ...Missing: cytogenetics | Show results with:cytogenetics
  54. [54]
    Points to consider in the detection of germline structural variants ...
    Research has demonstrated that mate-pair sequencing identifies >90% of cytogenetically visible BCRs, revises the breakpoint location by at least 1 sub-band ...
  55. [55]
    Mate-Pair Sequencing Enables Identification and Delineation of ...
    Jan 3, 2025 · Mate-pair sequencing detects both balanced and unbalanced structural variants (SVs) and simultaneously informs in relation to both genomic location and ...Missing: Next- Generation
  56. [56]
    Single-cell aneuploidy and chromosomal arm imbalances define ...
    Oct 31, 2025 · The scDNA-seq with its WGS approach provided ground truth results for genome-wide, single-cell resolved aneuploidy events, allelic arm ...
  57. [57]
    Mosaic autosomal aneuploidies are detectable from single-cell ...
    Nov 25, 2017 · We have developed a method that uses chromosome-wide expression imbalances to identify aneuploidies from single-cell RNA-seq data.
  58. [58]
    Single-cell RNA-seq reveals a distinct transcriptome signature ... - NIH
    We distinguished aneuploid cells from diploid cells within the hematopoietic stem and progenitor cells using scRNA-seq. Monosomy 7 cells showed downregulated ...Missing: scWGS | Show results with:scWGS
  59. [59]
    Manta: rapid detection of structural variants and indels for germline ...
    Manta is optimized for rapid germline and somatic analysis, calling structural variants, medium-sized indels and large insertions on standard compute hardware.
  60. [60]
    Comparison of structural variant callers for massive whole-genome ...
    Mar 28, 2024 · Here, we compared the performances of 11 SV callers: Delly, Manta, GridSS, Wham, Sniffles, Lumpy, SvABA, Canvas, CNVnator, MELT, and INSurVeyor.
  61. [61]
    The Emergence of Artificial Intelligence-Guided Karyotyping
    May 31, 2025 · P421: AI-assisted karyotyping improves efficiencies at scale in the cytogenetics laboratory. Genet. Med. Open. 2023;1:100468. doi: 10.1016/j ...Missing: 2020s | Show results with:2020s
  62. [62]
    Incorporating automation in a cytogenetics laboratory
    Jun 12, 2025 · Artificial intelligence (AI)-assisted karyotype analysis software. This type of software utilizes AI algorithms to identify chromosomes ...Missing: 2020s | Show results with:2020s
  63. [63]
    KaryoXpert: : An accurate chromosome segmentation and ...
    Jul 1, 2024 · Abstract. Automated karyotyping is of great importance for cytogenetic research, as it speeds up the process for cytogeneticists through ...
  64. [64]
    Genome Evolution Due to Allopolyploidization in Wheat - PMC
    The wheat group has evolved through allopolyploidization, namely, through hybridization among species from the plant genera Aegilops and Triticum followed by ...
  65. [65]
    The Origin and Behavior of Mutable Loci in Maize - PMC - NIH
    The origin and behavior of mutable loci in maize. Barbara McClintock. Barbara McClintock. 1. Department of Genetics, Carnegie Institution, Cold Spring Harbor, ...Missing: cytogenetics | Show results with:cytogenetics
  66. [66]
    Inversions in the Chromosomes of Drosophila Pseudoobscura - PMC
    Sturtevant A. H., Dobzhansky T. Inversions in the Third Chromosome of Wild Races of Drosophila Pseudoobscura, and Their Use in the Study of the History of the ...Missing: cytogenetics 1930s
  67. [67]
    Genetic, cytogenetic, and molecular analyses of mutations induced
    The original mutant also carried a translocation that has segregated independently of the d-locus mutation (the chromosomes involved in the translocation have ...
  68. [68]
    Genomic Structure and Evolution of the Ancestral Chromosome ...
    Human chromosome 2 was formed by the head-to-head fusion of two ancestral chromosomes that remained separate in other primates. Sequences that once resided ...
  69. [69]
    Multiple independent origins of sex chromosomes in amniotes - PNAS
    Nov 28, 2006 · Evidence for different origin of sex chromosomes in snakes, birds, and mammals and step-wise differentiation of snake sex chromosomes.
  70. [70]
    Genome-wide impacts of alien chromatin introgression on wheat ...
    Mar 16, 2020 · Linkage drag, caused by alien chromosome segments introduced into wheat genetic backgrounds, has led to unfavorable agronomic and end-use ...
  71. [71]
    The adaptive significance of chromosomal inversion polymorphisms ...
    Chromosomal inversions, structural mutations that reverse a segment of a chromosome, cause suppression of recombination in the heterozygous state.
  72. [72]
    Adaptive Role of Inversion Polymorphism of Drosophila subobscura ...
    Drosophila subobscura Collin (Diptera: Drosophilidae) possesses one of the richest inversion polymorphism in the genus, on all five acrocentric chromosomes with ...
  73. [73]
    An Ancestral Balanced Inversion Polymorphism Confers Global ...
    Jun 1, 2023 · In Drosophila melanogaster, the widespread inversion polymorphism In(3R)Payne underpins latitudinal clines in fitness traits on multiple ...
  74. [74]
    Chromosomal inversion polymorphisms and adaptation - PubMed
    In the fruit fly Drosophila, chromosomal polymorphisms were used in classic studies of natural selection. Recent molecular genetic studies suggest that ...
  75. [75]
    Unprecedented within-species chromosome number cline in the ...
    This cline is exceptional in the geographic area that it covers (6000 km) and in its range of within-species chromosome number variation (2n = 56-106).
  76. [76]
    Chromosomal Speciation Revisited: Modes of Diversification in ...
    Mar 18, 2011 · Chromosomal rearrangements can alter the rate and patterns of gene flow within or between species through a reduction in the fitness of ...
  77. [77]
    Meiotic drive mechanisms: lessons from Drosophila - PubMed Central
    Oct 23, 2019 · Meiotic drivers are selfish genetic elements that bias their transmission into gametes, often to the detriment of the rest of the genome.
  78. [78]
    The Selfish Segregation Distorter Gene Complex of Drosophila ...
    Segregation Distorter (SD) is an autosomal meiotic drive gene complex found worldwide in natural populations of Drosophila melanogaster.
  79. [79]
    Conservation Genetics of the Cheetah - PubMed Central - NIH
    The cheetah (Acinonyx jubatus) is one of the most recognized examples of the important links between evolutionary history, genetic variation, and conservation.
  80. [80]
    Extreme genomic erosion after recurrent demographic bottlenecks in ...
    Dec 14, 2016 · The Iberian lynx (Lynx pardinus) is the most endangered felid and a unique example of a species on the brink of extinction. ... The karyotype of ...
  81. [81]
    (PDF) A Study on Karyotype of the Asian Leopard Cat, Prionailurus ...
    Aug 6, 2025 · The cat has a highly conserved karyotype, closely resembling the ancestral karyotype ... cat species currently listed as vulnerable or endangered ...
  82. [82]
    Chromosomal inversion polymorphisms shape the genomic ... - Nature
    Oct 17, 2022 · These results show that inversion polymorphisms have a significant impact on recombination, genome structure and genetic diversity in deer mice.
  83. [83]
    Cytogenetic and Molecular Analyses of Philadelphia Chromosome ...
    The Ph is detected by G-band karyotyping in around 90% of CML patients among whom 5–10% may have variant chromosome types.
  84. [84]
    HER2 Testing in Breast Cancer - 2023 Guideline Update
    This evidence-based guideline update addresses uncommon clinical scenarios and improved clarity for infrequent HER2 test results of uncertain biologic or…
  85. [85]
    Genetics, Cytogenetics, and Epigenetics of Colorectal Cancer
    Chromosomal instability proceeds through two major mechanisms, missegregation that results in aneuploidy through the gain or loss of whole chromosomes, and ...2. Genetics Of Colorectal... · 3. Cytogenetics Of... · 4. Epigenetics And...
  86. [86]
    Hyperdiploidy: the longest known, most prevalent, and most ... - Nature
    Oct 20, 2022 · Hyperdiploidy is the largest genetic entity B-cell precursor acute lymphoblastic leukemia in children.Missing: seminal | Show results with:seminal
  87. [87]
    Karyotype complexity and prognosis in acute myeloid leukemia
    Jan 15, 2016 · A complex aberrant karyotype consisting of multiple unrelated cytogenetic abnormalities is associated with poor prognosis in patients with acute myeloid ...Patients And Methods · Results · Complex Karyotype With Three...
  88. [88]
    Clonal phylogeny and evolution of critical cytogenetic aberrations in ...
    Cytogenetic evolution was associated with inferior outcomes in MM. These findings suggest that QM-FISH is a valuable tool for the analysis of clonal phylogeny ...
  89. [89]
    Cytogenetics and Cytogenomics in Clinical Diagnostics - MDPI
    In conclusion, next-generation sequencing technologies, both short- and long-read, are integral to the modern cytogenomic diagnostic workflow. Their integration ...
  90. [90]
    [PDF] Molecular Cytogenetics: Bridging the Gap Between G-banding and ...
    Jul 29, 2024 · The transition from G-banding to next-generation sequencing represents a significant advancement in cytogenetics, offering unparalleled ...
  91. [91]
    Whole genome sequencing of cytogenetically balanced ...
    These findings can help explain the observation that ~37% of apparently balanced translocations actually present with imbalances and cryptic rearrangements ...
  92. [92]
    Sequencing 101: Structural variation - PacBio
    Dec 19, 2023 · Learn why structural variation is critical in human health and agriculture, and why HiFi sequencing is ideal for studying them.What Is Structural Variation... · Why Is Structural Variation... · Hifi Sequencing Offers An...Missing: molecule phasing 2020s
  93. [93]
    Mapping epigenetic modifications by sequencing technologies
    Sep 1, 2023 · ChIP–Seq is based on formaldehyde/paraformaldehyde-mediated protein-DNA crosslinking, followed by incubation with the specific antibodies to ...
  94. [94]
    Integration of Hi-C and ChIP-seq data reveals distinct types of ...
    Abstract. We have analyzed publicly available K562 Hi-C data, which enable genome-wide unbiased capturing of chromatin interactions, using a Mixture Poisso.
  95. [95]
    Molecular profiling for precision cancer therapies - Genome Medicine
    Jan 14, 2020 · This review summarizes the current and upcoming approaches to implement precision cancer medicine, highlighting the challenges and potential solutions.
  96. [96]
    Precision Oncology Guided by Genomic Profiling in Breast Cancer
    Jul 23, 2025 · This study contributes to the real-world evidence supporting the role of molecular tumor boards in providing personalized treatment to breast ...Missing: cytogenomic | Show results with:cytogenomic
  97. [97]
    SV - Mutational Signatures - COSMIC
    Structural variations (SVs) are large genomic changes typically exceeding 1kb in length, which impact the arrangement of the genome. Types of SVs include large ...Missing: cytogenetics | Show results with:cytogenetics
  98. [98]
    COSMIC: a curated database of somatic variants and clinical data ...
    Nov 1, 2023 · Characterising the genomic landscape of somatic mutations in cancer is a challenge that has been approached from several different directions.Missing: cytogenetic big
  99. [99]
    Challenges and Opportunities for Clinical Cytogenetics in the 21st ...
    Feb 15, 2023 · The powerful utilities of current DNA sequencing technology question the value of developing clinical cytogenetics any further.
  100. [100]
    Genome Sequencing as an Alternative to Cytogenetic Analysis in ...
    Mar 10, 2021 · We found that whole-genome sequencing provided rapid and accurate genomic profiling in patients with AML or MDS.Missing: decline | Show results with:decline
  101. [101]
    Structural variant detection in cancer genomes - Nature
    Mar 2, 2021 · Here, we explore current strategies for integrating SV callsets and to enable the use of tumor-specific SVs in precision oncology.Missing: database | Show results with:database<|separator|>
  102. [102]
    Low Pass Whole Genome sequencing - medRxiv
    May 22, 2025 · Points to consider in the detection of germline structural variants ... Low-pass whole genome sequencing is a reliable and cost-effective approach ...
  103. [103]
    Live genome imaging by CRISPR engineering: progress ... - Nature
    Jul 31, 2025 · CRISPR–Cas-based genome imaging often targets genomic regions with repetitive sequences such as centromeres, alpha satellites and telomeres, ...
  104. [104]
    Enhancing chromosomal analysis efficiency through deep learning ...
    May 30, 2024 · The AI analysis system significantly improves the speed of chromosome karyotype analysis, reduces manual workload, and enhances work efficiency.
  105. [105]
    The clinical application and challenges of preimplantation genetic ...
    PGT plays a pivotal role in the primary prevention of birth defects, mainly chromosomal abnormalities and monogenic disease with known pathogenic variants.
  106. [106]
    Overview of pharmacogenomics - UpToDate
    Aug 14, 2025 · Among the most commonly studied are genetic sequence variants, structural changes in chromosomes (eg, translocations), epigenetic variants ...
  107. [107]
    Ethical concerns of direct-to-consumer genetic testing
    Apr 4, 2023 · A recent study showed DTCGT may lead to high false-positive rates (40%) and misclassification of common benign genetic variants as “high risk.”.
  108. [108]
    'Anonymous' genetic databases vulnerable to privacy leaks - Nature
    Oct 14, 2024 · The ability to link private and public data sets could be putting research participants' private health information at risk.
  109. [109]
    Quantum Computing Models Chromatin Folding And Gene ...
    May 31, 2025 · Researchers are investigating the relationship between epigenetic modifications and three-dimensional chromatin organisation by employing quantum annealing.
  110. [110]
    The Central Role of Cytogenetics in Radiation Biology - Allen Press
    Jul 10, 2024 · Below we summarize the kinds of chromosomal aberrations (CAs) produced by ionizing radiation, the nomenclature adopted to describe them, the ...