Sport | Horgászat » Volpi-Bridger - Fish Glossary, An Overview of the Fluorescence in Situ Hybridization Technique

Alapadatok

Év, oldalszám:2008, 14 oldal

Nyelv:angol

Letöltések száma:2

Feltöltve:2018. június 18.

Méret:773 KB

Intézmény:
-

Megjegyzés:

Csatolmány:-

Letöltés PDF-ben:Kérlek jelentkezz be!



Értékelések

Nincs még értékelés. Legyél Te az első!


Tartalmi kivonat

Source: http://www.doksinet Review FISH glossary: an overview of the fluorescence in situ hybridization technique Emanuela V. Volpi1 and Joanna M Bridger2 BioTechniques 45:385-409 (October 2008) doi 10.2144/000112811 The introduction of FISH (fluorescence in situ hybridization) marked the beginning of a new era for the study of chromosome structure and function. As a combined molecular and cytological approach, the major advantage of this visually appealing technique resides in its unique ability to provide an intermediate degree of resolution between DNA analysis and chromosomal investigations while retaining information at the single-cell level. Used to support large-scale mapping and sequencing efforts related to the human genome project, FISH accuracy and versatility were subsequently capitalized on in biological and medical research, providing a wealth of diverse applications and FISH-based diagnostic assays. The diversification of the original FISH protocol into the impressive

number of procedures available these days has been promoted throughout the years by a number of interconnected factors: the improvement in sensitivity, specificity and resolution, together with the advances in the fields of fluorescence microscopy and digital imaging, and the growing availability of genomic and bioinformatic resources. By assembling in a glossary format many of the “acronymed” FISH applications published so far, this review intends to celebrate the ability of FISH to re-invent itself and thus remain at the forefront of biomedical research. INTRODUCTION The introduction of fluorescence in situ hybridization (FISH) almost 30 years ago marked the beginning of a new era for the study of chromosome structure and function. Conceptually, FISH is a very straightforward technique that essentially consists in hybridizing a DNA probe to its complementary sequence on chromosomal preparations previously fixed on slides. Probes are labeled either directly, by incorporation of

fluorescent nucleotides, or indirectly, by incorporation of reporter molecules that are subsequently detected by fluorescent antibodies or other affinity molecules. Probes and targets are finally visualized in situ by microscopy analysis. As a combined molecular and cytological approach, the major advantage of this visually appealing technique resides in its unique ability to provide an intermediate degree of resolution between DNA analysis and chromosomal investigations, while also retaining information at the single-cell level. FISH gained widespread recognition as a physical mapping technique to support large-scale mapping and sequencing efforts related to the human genome project; however, its accuracy and adaptability were simulta1University Vol. 45 ı No 4 ı 2008 neously, or soon after, exploited in other areas of biological and medical research. As a result, a wealth of diverse applications and FISH-based diagnostic assays have been developed within different fields of

investigation, including clinical genetics, neuroscience, reproductive medicine, toxicology, microbial ecology, evolutionary biology, comparative genomics, cellular genomics, and chromosome biology. The diversification of the original FISH protocol into the impressive number of procedures available these days has been promoted through the years by a number of interconnected factors, such as the improvement in sensitivity, specificity, and resolution of the technique, brought about by a better understanding of the chemical and physical properties of nucleic acids and chromatin, together with the advances in the fields of fluorescence microscopy and digital imaging, and the growing availability of genomic and bioinformatic resources. Here, we have assembled in a glossary format many of the FISH applications published so far. Although we intend this review to celebrate the versatility of this technique, it is of course impossible to cover every modification of FISH, and therefore we have

limited ourselves to variants that are named by combining a prefix with the acronym FISH. As seen in the many flavors of FISH described in the following, this flexibility has allowed the underlying technique to remain at the forefront of biomedical research over the last three decades. ACM-FISH ACM-FISH is a multicolor FISH assay for the simultaneous detection of numerical and structural chromosomal abnormalities in sperm cells (1). The abbreviation ACM refers to the concurrent hybridization of DNA probes for the alpha (centromere), classical (1q12), and midi (1p36.3) satellites of chromosome 1 for the specific detection of duplications and deletions of 1pter and 1cen, and for the identification of chromosomal breaks within the 1cen-1q12 region. The ACM technique originated from the integration of technical aspects and biological findings that emerged from previous FISH investigations of chromosome rearrangements in sperm (2), as well in other cell types (e.g, the assessment of breaks

in 1q12 in human lymphocytes) (3,4). Its application has led to significant of Oxford, Oxford and 2Brunel University, Uxbridge, UK www.biotechniquescom ı BioTechniques ı 385 Source: http://www.doksinet Review discoveries in the occurrence of chromosomal abnormalities in the sperm of healthy men, showing that spontaneous structural defects are more frequent than numerical ones and that chromosomal breaks preexist in human sperm before fertilization, and also providing evidence for reproducible donor differences in breakpoint locations within 1q12 (1). The assay has also been successfully used for the analysis of sperm of infertile men to show that oligozoospermic men carry a higher burden of transmissible chromosome damage; these results raising the question of possible elevated levels of chromosomal defects following intracytoplasmic sperm injections (ICSI) treatments (5). armFISH armFISH is a 42-color M-FISH variant (see below for M-FISH) that allows the detection of chromosomal

abnormalities at the resolution of chromosome arms (p- and q-arms of all 24 human chromosomes, except the p-arm of the Y and acrocentric chromosomes) (6). The protocol combines a commercially available M-FISH kit with an arm kit or a set of chromosome arm-specific painting probes (7). It is a straightforward, but significant, improvement to the standard M-FISH technique, the most obvious advantage being the increased resolution to the level of chromosome arms and the resulting ability to detect pericentric inversions. The assay has been successfully applied to reveal widespread chromosomal instability in glioma cell lines (8). CARD-FISH CARD-FISH, which stands for catalyzed reporter deposition-FISH, refers to the signal amplification obtained by peroxidase activity through the deposition of a large number of fluorescently labeled tyramine molecules in which the horseradish peroxidase (HRP)-labeled probe has bound (see also T-FISH). Improvements have been made to the technique to aid

the delineation of bacterial sequences, with one aspect of improvement being collecting bacteria on filters (9–11). There has also been a combination of CARD-FISH with microautoradiography that has been 386 ı BioTechniques ı www.biotechniquescom termed MICRO-CARD-FISH (12–14). Tritium is the radioisotope that is usually used and is taken up by active cells as 3H-aspartic acid. The filters on which the bacteria are captured are submitted to CARD-FISH, and they are then placed onto photographic emulsions. Cells can be assessed using a fluorescence microscope that has transmission light in addition. This allows both fluorescent, positive cells to be viewed by excitation of UV light and co-localization of silver grains by white light. combining the standard CB-FISH protocol with the 24-color SKY technology, since in one hybridization step, the DNA from any chromosome within the micronuclei can be identified (23,24) (see below for M-FISH and SKY). CO-FISH Cellular compartment

analysis of temporal (cat) activity by FISH is an ingenious experimental approach devised to investigate the dynamic interactions of neuronal populations associated with different behaviors or cognitive challenges (15). The method, based on RNA-FISH on cryosections followed by confocal analysis, was originally applied to study the environment-specific expression of the neural activity-regulated, immediateearly gene (IEG) Arc, and to monitor its cellular and subcellular distribution in the whole brain in rat (16). As a functional brain imaging technique, the uniqueness of catFISH resides in its ability to confer both temporal and cellular resolution to the analysis of gene expression patterns in brain, an important combination for the study of the dynamics of information processing. Chromosome orientation or CO-FISH is the name given to a FISH technique that uses single-stranded DNA probes to produce strand-specific hybridization. The technique relies on labeling by

5′-bromodeoxyuridine (BrdU) incorporation one strand of the sample cells’ DNA during S-phase. Metaphase chromosomes are prepared a number of hours after the BrdU pulse. Following Hoechst staining and UV irradiation, the newly synthesized DNA becomes nicked at the sites of BrdU incorporation. These nicks are enlarged using ExoIII, and the newly replicated DNA is removed, leaving the parental strand as a single-stranded template for the hybridization procedure. Initially, CO-FISH was designed to determine the orientation of tandem repeats within centromeric regions of chromosomes (25,26). This technique had also been useful in assessing aspects of translocated chromosomes, specifically Robertsonian (27,28), and also chromosomal inversions, since an inversion obviously changes the orientation of the involved chromosome segment (29,30). For a review on CO-FISH, see Bailey et al. (31) CB-FISH COBRA-FISH CB-FISH involves hybridization on binucleated cells in which cytokinesis has been

blocked by treatment with cytochalasin-B (CB). The term CB-FISH was coined by a research group investigating the mechanism by which the ratio of mosaic diploid cells in vivo increased in trisomy 21 cases (17). However, protocols combining FISH with the CB blocking assay for the cytological analysis of micronucleation and aneuploidy events had already appeared in a number of earlier molecular cytogenetic studies (18–21), including an investigation of chromosome 21 malsegregation in Alzheimer’s patients (22). Analysis of the chromosomal content of micronuclei can be facilitated by The prefix COBRA stands for combined binary ratio. This particular FISH protocol brings together combinatorial labeling with ratio labeling (32). The ratio labeling method allows different ratios of label to distinguish between probes. This permits the use of fewer fluorochromes to produce more pseudocolors, allowing the resolution of more than 24 colors within a specimen. The novel aspect of the COBRA

technique is that two sets of probes are ratio-labeled identically with three fluorochromes (usually two sets of 12 chromosomes for a human karyotype), but then one set is further labeled with an additional fourth fluorochrome. Indeed, a further fifth catFISH Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review fluorochrome can be used to make up to 48 color combinations for differential painting of human chromosome arms (33). The capacity to view more than 24 colors allows the delineation of additional sequences such as viral genome inserts or single-copy genes (34). For a comprehensive review of the technique see Raap and Tanke (35). COD-FISH Although COD-FISH is an abbreviation that has been used to describe three different hybridization techniques, the most common use is for chromosome orientation and direction-FISH. This protocol is similar to CO-FISH (see above), but when combined with the information about the directional organization of telomeric sequences, the

technique can be termed COD-FISH (30). COD-FISH can also stand for concomitant oncoprotein detection-FISH, a technique to visualize not only the loci signals for a particular oncogene, but also the protein product derived from this gene. The development of such a protocol was aimed at performing quantification of gene copy number and amount of protein product to help elucidate interesting mechanisms involved in the transcription and translation of a particular message (36). Others have called this type of regimen Immuno-FISH (see below), but the COD protocol does add a semiquantitative aspect to it. Tubbs and colleagues went on to improve their method using nanogold visualized with bright field microscopy (37). Another technique that has been termed COD-FISH is the combined CaCO3 optical detection-FISH, in which FISH is used to detect calcifying microrganisms in open ocean (38). COMBO-FISH COMBO-FISH is a method with no requirement for sample denaturation prior to hybridization. The

prefix COMBO stands for combinatorial oligonucleotide. The technique utilizes sequence information to identify regions of the genome where there are stretches of purines or pyrimidines and uses homopurine or homopyrimidine probes that are able to form triple helices with duplex genomic 388 ı BioTechniques ı www.biotechniquescom DNA (39). Homopurine or homopyrimidine regions of DNA 14 bp in length or more compose 1% to 2% of the human genome, with an average of 200 of such stretches in a 250-kb segment of the genome. Accordingly, specific probe sets can be constructed to target genomic regions of interest in that size range. The omission of the denaturation step makes the hybridization procedure less harsh on nuclear architecture, rendering this technique ideal for three-dimensional (3-D) analysis of genome organization (40,41). Comet-FISH Comet-FISH is a combination of the comet assay and FISH analysis. The comet assay, also called single-cell gel electrophoresis or the single-cell

gel test, is used to evaluate the amount of DNA breakage within single cells by running the DNA out of the nuclei into an agarose gel. The combined Comet-FISH method, consisting in releasing, by electrophoresis, the DNA onto agarose-coated microscope slides prior to in situ hybridization, allows specific sequences to be delineated in the comet head or tail, thus permitting the assessment of whether specific genomic regions are sensitive to DNA damage and breakage (42,43). With this technique, researchers have demonstrated that DNA damage susceptibility is associated with the gene density of a chromosome rather than the chromosome size (43). Further, damage to specific genes can be detected (44–47). The sensitivity of telomeres to damage has also been assessed successfully using this method (48). See Rapp et al. (49) for an overview of the technique Cryo-FISH Cryo-FISH is a promising in situ hybridization technique that makes use of ultrathin cryosections (150 nm thick) of well-fixed,

sucrose-embedded cells. The technique was recently devised and successfully applied to the study of spatial interrelationships of chromosome territories in the cell nucleus (50), a muchdebated aspect of chromosome organization in interphase. By combining a robust cell fixation procedure and ultracryomicrotome sectioning with two-dimensional (2-D) microscopy analysis (wide-field), this innovative technique maintains to preserve chromatin nanostructure while simultaneously presenting with a better efficiency of hybridization and resolution than canonical 3-D FISH, and provides an alternative, and possibly more userfriendly approach, to the study of genome organization in the nuclear context. Cryo-FISH was also recently used to validate results obtained by chromosome conformation capture on chip (4C) technology to demonstrate long-range chromosomal interactions of functional significance (51). D-FISH D-FISH is an enhanced version of the fusion signal-FISH protocol for the detection of

recurring chromosomal translocations in hematological malignancies (see below for fusion-signal FISH). The prefix D stands for double fusion, since in this particular protocol the use of two (or two sets of) differentially labeled, large probes, each spanning one of the two translocation breakpoints, allows the simultaneous visualization of both fusion products, significantly reducing the impact of false-negative results, a reason of concern in single fusion FISH. D-FISH was initially devised to improve detection of the double BCR/ABL fusion in chronic myeloid leukemia (CML) patients (52,53). Subsequently, D-FISH probes were developed for the 8;21 translocation and used for the assessment of minimal residual disease in acute myeloid leukemia (AML) patients during remission (54) and also for the visualization of PML/RARA double fusion in acute promyelocytic leukemia (APL) (55,56), the DEK/CAN double fusion resulting from t(6;9) in AML (57), and the PBX1/E2A double fusion in pediatric

patients with acute lymphoblastic leukemia (ALL) (58). A wide range of probes for D-FISH, as well as FusionSignal and Split-Signal FISH for the chromosomal analysis of hematological cancer, are now commercially available. DBD-FISH DBD-FISH stands for DNA breakage detection FISH, a technique developed Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review by Gosálvez and colleagues (59,60). Basically, this adaptation of the FISH procedure permits any sites of DNA damage/breakage in the sample genome to be analyzed in situ by means of an alkali DNA unwinding solution and protein removal. Cells are normally stabilized in agarose beads, but the technique can be applied to DNA comets (61). The incubation with the unwinding buffer leads to the presence of single-stranded DNA in the sample that can be hybridized with the appropriate probes (62,63). The technique has been used successfully to test the DNA fragmentation levels in sperm samples (64). e-FISH e-FISH is a BLAST-based FISH

simulation program able to accurately predict the outcome of hybridization experiments. The program was developed as one of the bioinformatics resources to be available from the Database of Genomic Variants, aimed at simplifying the choice of appropriate genomic probes for hybridization experiments and facilitating the interpretation of the results (65). This virtual FISH approach includes a repeat masking step mimicking in silico the COT-1 blocking of repetitive sequences. The program is freely accessible at projectstcagca/efish Fiber-FISH Fiber-FISH is a technique that allows high resolution mapping of genes and chromosomal regions on fibers of chromatin or DNA, permitting physical ordering of DNA probes down to a resolution of 1000 bp, and that also allows assessment of gaps and overlaps in contigs and analysis of segmental duplications and copy number variants. In practice, the method consists of releasing chromatin/DNA fibers from cell nuclei, usually by means of salt or solvent

extraction, and stretching and fixing them on a microscope slide prior to hybridization. Similar variants of the technique, in which the chromatin is run down the slide and is stretched by fluid flow, were initially set up more or less concomitantly by different research groups (66–68). However, the specific Fiber-FISH termi390 ı BioTechniques ı www.biotechniquescom nology was introduced slightly later (69,70). A significant improvement in terms of DNA stretching uniformity and reproducibility was provided by the implementation of the molecular combing protocol (71), which uses the action of a receding air/water meniscus to extend and align DNA molecules attached at one end to a glass surface. Flow-FISH In the Flow-FISH technique, as described by Lansdorp and colleagues in 1998 (72), PNA-labeled telomere probes are used to visualize and measure the length of telomere repeats, as in the Quantitative-FISH technique (see below for Q-FISH and PNA-FISH), but the analysis combines in

situ hybridization with flow cytometry for measurement of the telomeric signals from cells in suspension. This permits large numbers of cells to be analyzed rapidly. Lansdorp and his collaborators have developed this technique, and his and other laboratories have used it for a number of different cell types and clinical applications (73). Indeed, Flow-FISH has been used in aging studies (74–76), telomere maintenance (77,78), and in clinical applications for ex vivo suspension cells (hematopoietic) (79–84). Also, combining telomere Flow-FISH with fluorescent immunodetection of cell surface markers has advanced the understanding of the behavior of stem cell populations (80,85–88). Researchers have expanded the Flow-FISH technique to permit assessment of different strains of bacteria (89,90). Fusion-Signal FISH The Fusion-Signal FISH technique was initially devised for the identification of the 9;22 Philadelphia translocation in peripheral blood and bone marrow cells of CML patients

to detect minimal residual disease after bone marrow transplantation (91). BCR and ABL gene fragments, each flanking one of the two breakpoints, were used as probes for the detection of the BCR/ABL fusion product, hence the fusion-signal appellation. Since then, sets of probes to detect fused gene signals originating from a range of critical translocation events in hematological malignancies, for instance the PML and RARA fusion product resulting from t(15:17) in APL (92), have been designed by different research groups. With FISH assays available for CML and APL, as well as AML (93,94), non-Hodgkin’s lymphoma (95,96), mantle cell lymphoma (97), childhood B-lineage acute lymphoblastic leukemia (98–100), and infant leukemia (101), it would be difficult to deny the major impact of the fusion signal technique as a diagnostic and prognostic tool for blood cancer. However, soon after its conception, concerns started to emerge that the interpretation of the results was complicated by

the variable occurrence of false-positive and false-negative signals (102–104). To overcome these difficulties, an ingenious dual-fusion variant of the technique was devised for the detection of the 9;22 translocation in CML (52). The protocol involves the use of large probes, spanning the two breakpoints, for the simultaneous visualization of both fusion signals BCR/ABL and ABL/BCR. This modified and improved version of the fusion signal technique was named D-FISH (see above). Halo-FISH Halo-FISH is an acronym that describes FISH performed on cells that are first permeabilized and then extracted with high salt to remove soluble proteins (105,106). Indeed, chromatin/DNA that is not fixed to an internal structure within cell nuclei is released, forming a halo around a residual nucleus. FISH can then be performed on these preparations using any type of probe to delineate specific DNA sequences. Researchers have used α-satellite (106), telomeres (107), scaffold attachment regions

(SARs) (108), matrix attachment regions (MARs) (109), gene loci (105,110,111), and whole chromosomes (112,113). DNA halo preparations can be used for high-resolution mapping, since such long extended loops of DNA are created (114). A number of groups have used DNA halo preparations to analyze sperm chromatin, as it makes it easier to access sperm DNA, which is normally very compact due to its association with protoamines. Some have even described this type of analysis as SpermHalo-FISH (115), whereby sperm Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review nuclei are spun onto glass microscope slides and treated with dithiothreitol for permeabilization followed by high salt. Harlequin-FISH Harlequin-FISH is a method for cell cycle-controlled chromosome analysis in human lymphocytes that allows a precise quantification of induced chromosome damage for human biodosimetry purposes (116,117). The approach combines FISH painting with differential replication staining after BrdU

treatment of lymphocyte cultures (or harlequin staining). The principle, on which differential replication staining is based, is that after two rounds of replication in the presence of the base analog BrdU, sister chromatids will stain differentially (with either Giemsa and/or fluorescent dyes) (118,119). This allows the identification of the two chromatids and the observation of sister chromatid exchanges (SCEs), that after a few cell divisions confer to the chromosomes an asymmetrically striped appearance, to which the term harlequin refers. Since for accurate cytogenetic measurements of genetic damage, cells must be analyzed in their first mitosis following exposure, the most relevant aspect of the harlequin technique in combination with FISH for biodosimetry studies is that, according to sister chromatid staining patterns, cells in different division cycles can be distinguished, allowing chromosomal analysis to be carried out selectively. Immuno-FISH Immuno-FISH is a combination of

two techniques, one being standard FISH, either on flattened chromosome preparations (2-D FISH) or on three-dimensionally preserved nuclei (3-D FISH), and the other indirect or direct immunofluorescence. The latter technique permits the visualization of antigens within the sample, so that both DNA and proteins can be analyzed on the same sample. It is often used to investigate co-localization of genomic regions with proteinaceous entities within interphase nuclei such as nucleoli (120) or promyelocytic leukemia (PML) bodies (121). Not all antigens will be preserved after the various steps in the 392 ı BioTechniques ı www.biotechniquescom FISH protocols, but it is possible to apply the primary or primary and secondary antibodies and proceed with a fixation step prior to the FISH procedure (122). The term Immuno-FISH was first coined by Brown et al. in 1997 (123), in which co-localization of active or inactive gene loci were assessed with nuclear structures containing the Ikaros

protein, although others had combined the two methodologies previously for RNA-FISH with splicing speckles (124) and for anti-CENP C staining in combination with α-satellite probes (125). The combination of these techniques has been used for great effect, even helping to position chromosomes in interphase nuclei (112), and is now being used with multiple probes and colors, such as in the paper from Cremer’s group, whereby they look at chromosomal regions, gene expression, and histone methylation (126). LNA-FISH Locked nucleic acids (LNAs) are a class of RNA analogs with exceptionally high affinity toward complementary DNA and RNA (127). Because of the LNA chemical makeup, heteroduplexes between LNA oligonucleotides and their complementary DNA oligonucleotides show a shift in structure from a B-like helix toward an A-type helix. This results in a higher thermal stability of the LNA-DNA heteroduplexes (128). LNA-FISH refers to the use of LNA-modified oligonucleotides in FISH

experiments for improved resolution and sensitivity (129,130). M-FISH The invention of M-FISH (or Multiplex-FISH), a protocol for 24-color karyotyping, based on combinatorial labeling (131) and aimed at facilitating the analysis of complex chromosomal rearrangements and marker chromosomes, has signified a groundbreaking development in molecular cytogenetics, particularly for the study of tumors and prenatal diagnosis. Dissimilarly from ratio-labeling based multicolor approaches, in which chromosomespecific probes are characteristically labeled with different proportions of fluorochromes and accurate measure- ments of relative fluorescence intensity are required, the M-FISH technique (like the related SKY technique, see below) consists of labeling each probe with a unique combination of five spectrally separable fluorochromes in a 1:1 ratio. Accordingly, although relying on the use of narrow band-pass fluorescence filters appropriately set in a motorized filter wheel and on digital

imaging software more sophisticated than the standard setup for FISH analysis, the interpretation of the results is relatively straightforward (that is, the fluorochrome is either present or absent). The technique was originally devised for use with and simultaneous detection of the 24 human chromosome painting probes (22 autosomes and the X and Y chromosomes), but has been subsequently used to analyze specific chromosomal subregions, like centromeres and subcentromeres in protocol variants for the characterization of small supernumerary marker chromosomes with no euchromatin. Examples include cenM-FISH (132), CM-FISH (133), and subcenM-FISH (134), telomeres for the identification of subtle subtelomeric rearrangements as in M-TEL (135,136), and chromosome arm-specific probes (see armFISH above) for the detection of pericentric inversions. Similar in principle and application to the M-FISH technique is the spectral karyotyping technique or SKY (137), in which chromosomes are classified

on the basis of their unique emission spectra. For image acquisition and analysis, SKY requires specific hardware and software, comprising a custom-designed single triple-band-pass filter and an interferometer able to retrieve spectral information for every pixel in a digital image. For comprehensive reviews on 24-color FISH analysis see Kearney et al. (2006) (138) and Schrock et al (2006) (139). M-FISH is also used as a term to mean Multicolor-FISH. Multilocus or ML-FISH The word multilocus (subsequently abbreviated with the acronym ML) refers to the simultaneous use in multicolor FISH of multiple probes. This FISH assay was initially designed to screen for multiple microdeletion syndromes in patients with unexplained developmental delay and/or mental retardation (140). Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review The original multilocus panel designed included Prader-Willi, Angelman, Williams, Di George/velocardiofacial, and Smith-Magenis syndromes. Based on the same

principle, a multilocus strategy with locus-specific DNA probes for chromosomes 13 and 21 was more recently applied to the study of nondisjunction in mature human oocytes (141). PCC-FISH PCC-FISH is a FISH application used for biodosimetric analysis that relies on the use of chromosome-specific painting probes to determine chromosome damage after irradiation. The acronym PCC stands for premature chromosome condensation and refers to the effect obtained by virusmediated cell fusion or phosphatase inhibitors (either calyculin A or okadaic acid) to prematurely condense the chromosomes of cells in G1 and G2 phases, a central aspect of the procedure that overcomes the need to culture cells in vitro as required for conventional metaphase chromosome analysis. PCC-FISH was initially devised as an assay to estimate/ predict the in situ radiation sensitivity of individual human tumors (142–144). It has subsequently been used to estimate the effect of whole-body high- or low-dose exposure to

human peripheral lymphocytes (145,146), and carried out on skin fibroblasts in the case of acute localized irradiation following accidental overexposure (147). PCC-FISH has also been used to establish the radiosensitizing effect of drugs in experimental cancer treatments (75,148,149). The technique is used additionally in in vitro studies for general radiation research and cancer research purposes. PNA-FISH Peptide nucleic acids (PNAs) are synthetic analogs of DNA in which the deoxyribose phosphate backbone supporting the nucleic acid bases is replaced by a noncharged peptide backbone (150). As a result of this unique structural property, there is no electrostatic repulsion when PNA oligomers hybridize to complementary DNA or RNA sequences, and PNA-DNA and PNA-RNA duplexes are more stable than 394 ı BioTechniques ı www.biotechniquescom the natural homo- or heteroduplexes. FISH with PNA probes or PNA-FISH was first used by Landsdorp and collaborators (151) to measure individual

telomere lengths on metaphase chromosomes. Quantitative telomere analysis by PNA-FISH was subsequently carried out on interphase cells (152). The discovery that PNA-DNA hybridization is more significantly affected by base mismatches than DNA-DNA hybridization (153) and that PNA probes could discriminate between two centromeric repeats that differed only by a single base pair (154) endorsed this as an important development in the field. Thus, short (15- to 18-mer) PNA probes for α-satellite domains of specific chromosomes were designed, and their power of discrimination at a single-base level was used for unique chromosome identification in metaphase and interphase (155,156). As well as on lymphocytes, amniocytes and fibroblasts, PNA-FISH has also been successfully carried out on human spermatozoa (157) and isolated oocytes, polar bodies, and blastomeres (158), a strong indication of the potential of PNA probes for preimplantation cytogenetic diagnosis. Larsen and collaborators (159)

tested the suitability of PNA-FISH for noninvasive prenatal diagnosis by detecting γ-globin γ messenger RNA (mRNA) in fetal nucleated red blood cells from maternal blood. Also, as a result of the relative hydrophobic character of PNA compared with DNA, that allows better diffusion through the cell wall, PNA-FISH has also had wide application in microbiology, for both research and clinical purposes (160,161). Because of its high binding specificity quality, PNA technology is expected to provide a platform for the design of allele-specific probes for in situ hybridization, a technical development longed for by many in the genetic, cytogenetic, and epigenetic ranks. Q-FISH Quantitative-FISH methodology permits the measurement of probe signal intensity. Q-FISH was initially invented by Lansdorp and collaborators (162) and was shown to work in combination with flow cytometry (163) (see above for Flow-FISH). This method has been used mainly for measuring the number of telomere repeats on

a particular chromosome, using PNA-conjugated probes. Typically, metaphases are imaged and then analyzed using software such as TFL-TELO (162,164). The lengths of chromosomal regions can be measured at the resolution of 200 bp (165). Q-FISH has advanced and is now used on interphase cells and tissue sections. Indeed, in interphase cells, a similar methodology with a different acronym has been used (i.e, single telomere length analysis or STELA) (166). Q-FISH has also been used to quantify telomere length in interphase cells (167) and has even been given the acronym IQ-FISH for Interphase Q-FISH (168). Q-FISH has become an important tool in studying the role of telomeres in aging and cancer. QD-FISH QD-FISH refers to the recently pioneered utilization of quantum dot-conjugates in FISH protocols. Quantum dots are nanometer-sized inorganic fluorophores, characterized by photostability and narrow emission spectra, that have been successfully used for FISH analysis on human metaphase

chromosomes (169), human sperm cells (170), bacterial cells (171), and also to detect subcellular mRNA distribution in tissue sections (172). While initial QD-FISH protocols entailed hybridization of biotinylated probes and subsequent detection using streptavidin-conjugated QDs, more recent adaptations of the procedure, aimed at multicolor imaging, involve direct labeling of oligonucleotide probes (172,173). Rainbow-FISH Rainbow-FISH is an advanced digital imaging procedure that allows the simultaneous detection and quantification of up to seven different microbial groups in a microscopic field. The technique was designed to improve quantitative analytical studies of microbial communities from various aquatic environments (174). Based on similar principles to the multi-FISH technique previously developed for the analysis of microbes in human feces (175), the rainbow protocol brings together the use of specific 16S Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review ribosomal

RNA (rRNA)-targeted oligonucleotide probes for the discrimination of different phylogenetic groups of microbes (176) and the principles of combinatorial labeling. As a result, by the combined application of seven DNA probes, each labeled with up to three fluorochromes, seven kinds of microbial strains can be distinguished simultaneously (177). The specific Rainbow-FISH digital procedure developed by Sunamura and Maruymama (174) consists of systematic background noise reduction and target signal equalization to discriminate between microbes and nonmicrobial particles, and aims at facilitating image processing and analysis, in particular the normalization steps that are usually very laborious when imaging natural environmental microbes, since their cellular rRNA content can vary between cells, even within the same sample. Raman-FISH Raman-FISH is a new technique that combines FISH technology with Raman microspectroscopy for ecophysiological investigations of complex microbial communities

(178). The procedure exploits the so-called red shift phenomenon or the significant change in the resonance spectra, as visualized by Raman microscopy, that follows anabolic incorporation of 13C isotope, when compared with normal 12C, into microbial cells. Metabolic labeling through stableisotope incorporation is combined with microbial species identification by in situ hybridization with specific 16S rRNA probe for structural and functional interrelated analyses of microbial communities at a single-cell resolution. ReD-FISH ReD-FISH, which stands for replicative detargeting FISH, is similar to CO-FISH, whereby BrdU is added to the culture medium during DNA replication for incorporation into the newly synthesized strand. ReD-FISH allows the replication timing of specific sequences to be determined. If BrdU has been incorporated in the sequence of interest, the newly formed DNA strand will be detargeted (as in CO-FISH, see above), 396 ı BioTechniques ı www.biotechniquescom and each

oligonucleotide probe will only be able to hybridize to one of the parental strands, and only one chromatid will be display a signal. However, if the sequence of interest has not been replicated and not incorporated BrdU, then a FISH analysis will reveal the standard double signal on both chromatids of the metaphase chromosome. The ReD-FISH technique has been instrumental in ascertaining the replication timing of telomeres, leading to the observation that telomeres replicate throughout S-phase and that telomeres from p and q arms of the same chromosome replicate asynchronously (179). Reverse-FISH First demonstrated in 1990 (180), Reverse-FISH is the process whereby the FISH probe comprises DNA from the material of interest. This can be a chromosome of a specific species in the cellular background of another species (i.e, a somatic cell hybrid) It can also be chromosomal material obtained in other ways such as by flow sorting (181) or microdissection (182,183). The reverse terminology

refers to the probe being the material of interest, usually aberrant, and being painted onto control or reference metaphase chromosomes to identify what sequences/chromosomal regions the probe contains or is missing. Reverse-FISH has been useful for characterizing marker chromosomes (184–186) and chromosome amplifications in cancer (187). RING-FISH In situ hybridization of oligonucleotide probes to high copy number nucleic acid targets, such as rRNA, is a standard method for the identification of microorganisms in enviromental samples. RING-FISH is a modified version of this technique that relies on the use of high concentrations of polynucleotide probes for an increase in sensitivity and visualization of any part of the genetic material of a bacterial cell, regardless of copy number (188,189). The signal amplification achieved with these polynucleotides, the length of which ranges between one to several hundred base pairs, is mediated by their secondary structures and intermolecule

interactions, resulting in a conspicuous network of polynucleotides that builds upon the initial hybridization site. RING stands for recognition of individual genes, but also refers to the characteristic ring-shaped or halo appearance of the fluorescence signals at the bacterial cell periphery (188). RNA-FISH RNA-FISH is a method that allows detection of RNA within cells. Transcripts can be visualized either in the nucleus or in the cytoplasm. The technique, also known as expression-FISH, has been used to analyze the transcriptional activity of endogenous genes (190) as well as exogenous genes such as those belonging to integrated viral genomes (191,192) and transgenes (193). The technique permits investigation into allelic-specific expression on a per cell basis (194,195) and is expected to provide a platform for gene expression profiling studies in single cells (196). RNA-FISH has also been instrumental in studying different functional aspects of genome organization and nuclear

architecture (124,197– 205). Further, as a technique, it is being examined as a prenatal diagnosis tool for myotonic dystrophy type 1 (206). RxFISH RxFISH is a color banding technique that is also described as chromosome bar-coding (207). The method relies on sequence homologies between human and the apes, such as gibbon (98%), to produce, by cross-species hybridization, a specific banding pattern on human metaphase chromosomes (208–210). This is possible because of the intrachromosomal rearrangements that have occurred during primate evolution. If the probes are labeled with a number of fluorochromes, usually three, this allows a colorful and reproducible banding to be observed and analyzed. Due to the color bands, it is easier than G-banding to see chromosomal rearrangements, especially intrachromosomal rearrangements (211). However, in combination with G-banding, RxFISH can provide very detailed information about chromosomal breakpoints, for example in cancer (212–214). Vol.

45 ı No 4 ı 2008 Source: http://www.doksinet Review Split-Signal FISH Split-Signal FISH is a fast and sensitive dual-color FISH assay for the detection of frequently occurring chromosome translocations affecting specific genes in hematopoietic malignancies. The technique is equally suitable for metaphase and interphase analysis and has been increasingly used for both diagnostic and prognostic purposes. In its current form, the assay involves the design and differential labeling of two probes from the flanking regions of the translocation breakpoint. The signals normally co-localize and appear fused, but as a result of the translocative event, they will split. Split-Signal FISH was initially introduced as an innovative and simple experimental approach for the detection of all types of MLL gene translocations in ALL and AML, using only a single FISH test (215). However, a prototypic version of the protocol with differentially labeled probes for genes spanning or adjacent to the

translocation breakpoints, on the two chromosomes involved, had been previously used for the detection of the Burkitt translocation t(8;14) in B cell lymphomas (216) and the detection of t(11;14) in mantle cell lymphomas (217). See also related Fusion-Signal FISH technique. T-FISH The T in T-FISH can stand for tyramide, tissue, or telomere. The three versions of T-FISH are discussed in the order of their arrival in the field. Tyramide-FISH: tyramide is a compound that binds to peroxidase easily and thus has been used to increase the sensitivity greatly in FISH experiments, with the use of only one or two layers of reagents for visualization (218). The first layer uses a peroxidase-conjugated antihapten antibody or a compound such as strepavidin to bind to the labeled probe. Fluorochromes or haptens, such as biotin, are conjugated to tyramine derivatives. This leads to a massive build-up or towers of fluorochromes or moieties that can be visualized by fluorochromes, making detection

ultrasensitive (219). The chemistry behind tyramide signal amplification (TSA) is now licensed, and kits can be purchased from various companies. The technology has been used to map 398 ı BioTechniques ı www.biotechniquescom gene loci and look for specific transcripts in cells (220–222). Tissue-FISH: tissue samples are collected frequently from patients or experimental animals. These samples can be frozen, fixed, or embedded in paraffin wax. A number of research groups have developed methods of delineating sequences in tissues by FISH, for example, in paraffin sections and frozen sections (223). However, it was Nomura and collaborators (224), who, in 2003, coined the term T-FISH to denote FISH on tissue sections. Telomere-FISH: some groups have called FISH using telomeric probes T-FISH (225–227). 3-D FISH 3-D FISH was developed in Germany by the groups of Peter Lichter and Thomas Cremer (124,228,229) to analyze spatial positioning and relative organization of chromosomes and

subchromosomal regions within cell nuclei. The methodology relies on using cross-linking fixation reagents, such as paraformaldehyde, to preserve nuclear architecture and large-scale chromatin organization (230,231). Due to the resulting crosslinking of proteins, an effective permeabilization step is required to allow the probes to penetrate the sample. This is often performed using detergents and freeze and thaw cycles at -180°C. To visualize the 3-D signals, operators will need to use confocal laser-scanning microscopy or use accurate deconvolution tools. One of the most significant achievements in the field was the publication of a paper displaying the simultaneous painting of all human chromosomes in the nuclei of three-dimensionally preserved cells (232). The introduction of 3-D FISH protocols and their wide application in the field of chromosome biology has significantly propelled, over the last few years, our understanding of chromosome structure and function. Zoo-FISH

Zoo-FISH, also known as crossspecies chromosome painting, consists of hybridizing libraries of DNA sequences, also known as chromosome paints, from one species to the chromosomes of another species, to identify regions of synteny. From a methodological point of view, the protocol does not differ significantly from standard FISH protocols, but there can be some issues with signal intensity and background noise, as pointed out by Solovei (www.biologie uni-muenchen.de/ou/humbio/pdf/ solovei/protocol/11 zoofish.pdf) Solovei discusses that good metaphase spreads are absolutely fundamental, that suppression of repetitve sequences may be unnecessary, and that higher concentrations of probe are usually required. A critical step to be borne in mind is that the more divergent the organisms are from each other, the lower the stringency should be within the protocol. The first study using Zoo-FISH used human and mouse whole chromosome painting probes on primates, rodents, even-toed ungulates, and

whales (233). Since then, this type of chromosomal analysis has been widely used, permitting the establishment of important chromosome homology maps, revealing interesting insights into chromosome rearrangements during evolution, and allowing us to infer information on ancestral karyotypes. The impact of Zoo-FISH on the comparative study of genome evolution has been reviewed in different papers (234–237). ACKNOWLEDGMENTS The development of such an abundance of exciting FISH protocol variants and applications that we report and discuss in this glossary is the result of numerous, generally independent, but also collaborative efforts of many different laboratories in the world. Due to space limitations we have been unable to cite all relevant/significant papers, and we would like to apologize to friends and colleagues whose contribution to the field has been unwillingly omitted. COMPETING INTERESTS STATEMENT The authors declare no competing interests. REFERENCES 1. Sloter, ED, X Lowe,

ID Moore, J Nath, and A.J Wyrobek 2000 Multicolor FISH analysis Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review of chromosomal breaks, duplications, deletions, and numerical abnormalities in the sperm of healthy men. Am J Hum Genet 67:862-872 2. Van Hummelen, P, XR Lowe, and AJ Wyrobek. 1996 Simultaneous detection of structural and numerical chromosome abnormalities in sperm of healthy men by multicolor fluorescence in situ hybridization. Hum Genet 98:608-615. 3. Eastmond, DA, DS Rupa, and LS Hasegawa. 1994 Detection of hyperdiploidy and chromosome breakage in interphase human lymphocytes following exposure to the benzene metabolite hydroquinone using multicolor fluorescence in situ hybridization with DNA probes. Mutat. Res 322:9-20 4. Rupa, DS, L Hasegawa, and DA Eastmond 1995. Detection of chromosomal breakage in the 1cen-1q12 region of interphase human lymphocytes using multicolor fluorescence in situ hybridization with tandem DNA probes. Cancer Res. 55:640-645 5.

Schmid, TE, MH Brinkworth, F Hill, E. Sloter, A Kamischke, F Marchetti, E Nieschlag, and A.J Wyrobek 2004 Detection of structural and numerical chromosomal abnormalities by ACM-FISH analysis in sperm of oligozoospermic infertility patients. Hum Reprod 19:1395-1400. 6. Karhu, R, M Ahlstedt-Soini, M Bittner, P. Meltzer, JM Trent, and JJ Isola 2001 Chromosome arm-specific multicolor FISH. Genes Chromosomes Cancer 30:105-109. 7. Guan, XY, H Zhang, M Bittner, Y Jiang, P Meltzer, and J. Trent 1996 Chromosome arm painting probes. Nat Genet 12:10-11 8. Sallinen, SL, P Sallinen, M Ahlstedt-Soini, H. Haapasalo, H Helin, J Isola, and R Karhu. 2003 Arm-specific multicolor fluorescence in situ hybridization reveals widespread chromosomal instability in glioma cell lines. Cancer Genet. Cytogenet 144:52-60 9. Pernthaler, A, J Pernthaler, and R Amann 2002. Fluorescence in situ hybridization and catalyzed reporter deposition for the identification of marine bacteria. Appl Environ Microbiol

68:3094-3101. 10. Sekar, R, A Pernthaler, J Pernthaler, F Warnecke, T. Posch, and R Amann 2003 An improved protocol for quantification of freshwater Actinobacteria by fluorescence in situ hybridization. Appl Environ Microbiol 69:2928-2935. 11. Tujula, NA, C Holmstrom, M Mussmann, R. Amann, S Kjelleberg, and G.R Crocetti 2006 A CARD-FISH protocol for the identification and enumeration of epiphytic bacteria on marine algae. J Microbiol. Methods 65:604-607 12. Nielsen, JL, C Klausen, PH Nielsen, M. Burford, and NO Jorgensen 2006 Detection of activity among uncultured Actinobacteria in a drinking water reservoir. FEMS Microbiol. Ecol 55:432-438 13. Sintes, E and GJ Herndl 2006 Quantifying substrate uptake by individual cells of marine bacterioplankton by catalyzed reporter deposition fluorescence in situ hybridization combined with microautoradiography. Appl Environ. Microbiol 72:7022-7028 14. Teira, E, T Reinthaler, A Pernthaler, J. Pernthaler, and GJ Herndl 2004 400 ı BioTechniques ı

www.biotechniquescom Combining catalyzed reporter deposition-fluorescence in situ hybridization and microautoradiography to detect substrate utilization by bacteria and Archaea in the deep ocean. Appl Environ. Microbiol 70:4411-4414 15. Guzowski, JF, BL McNaughton, CA Barnes, and P.F Worley 2001 Imaging neural activity with temporal and cellular resolution using FISH. Curr Opin Neurobiol 11:579-584. 16. Guzowski, JF, BL McNaughton, CA Barnes, and P.F Worley 1999 Environmentspecific expression of the immediate-early gene Arc in hippocampal neuronal ensembles. Nat Neurosci. 2:1120-1124 17. Chen, J, Q Shi, J Zhang, H Li, X Zhang, J. Luo, G Zhou, X Shan, and X Zhang 2000. Detection of mosaic chromosome 21 aneuploidy in vivo with CB-FISH method. Zhonghua Yi Xue Yi Chuan Xue Za Zhi 17:196-199. 18. Catalan, J, K Autio, E Kuosma, and H Norppa. 1998 Age-dependent inclusion of sex chromosomes in lymphocyte micronuclei of man. Am J Hum Genet 63:1464-1472 19. Kirsch-Volders, M, I Tallon, C

Tanzarella, A. Sgura, T Hermine, EM Parry, and JM Parry. 1996 Mitotic non-disjunction as a mechanism for in vitro aneuploidy induction by X-rays in primary human cells. Mutagenesis 11:307-313. 20. Minissi, S, B Gustavino, F Degrassi, C Tanzarella, and M. Rizzoni 1999 Effect of cytochalasin B on the induction of chromosome missegregation by colchicine at low concentrations in human lymphocytes. Mutagenesis 14:43-49. 21. Surralles, J, G Falck, and H Norppa 1996. In vivo cytogenetic damage revealed by FISH analysis of micronuclei in uncultured human T lymphocytes. Cytogenet Cell Genet. 75:151-154 22. Migliore, L, N Botto, R Scarpato, L Petrozzi, G. Cipriani, and U Bonuccelli 1999. Preferential occurrence of chromosome 21 malsegregation in peripheral blood lymphocytes of Alzheimer disease patients. Cytogenet Cell Genet. 87:41-46 23. Komae, N, Y Hibino, and N Sugano 1999 [Analysis of micronuclei induced under hyperthermic conditions in human lymphocyte culture by fluorescence in situ

hybridization (FISH) and spectral karyotyping (SKY) methods.] [Article in Japanese] Yakugaku Zasshi 119:763-772. 24. Leach, NT and C Jackson-Cook 2001 The application of spectral karyotyping (SKY) and fluorescent in situ hybridization (FISH) technology to determine the chromosomal content(s) of micronuclei. Mutat Res. 495:11-19 25. Goodwin, E and J Meyne 1993 Strandspecific FISH reveals orientation of chromosome 18 alphoid DNA Cytogenet Cell Genet 63:126-127. 26. Meyne, J, EH Goodwin, and RK Moyzis 1994. Chromosome localization and orientation of the simple sequence repeat of human satellite I DNA. Chromosoma 103:99-103 27. Garagna, S, D Broccoli, CA Redi, JB Searle, H.J Cooke, and E Capanna 1995 Robertsonian metacentrics of the house mouse lose telomeric sequences but retain some minor satellite DNA in the pericentromeric area. Chromosoma 103:685-692. 28. Garagna, S, N Marziliano, M Zuccotti, J.B Searle, E Capanna, and CA Redi 2001. Pericentromeric organization at the fusion point of

mouse Robertsonian translocation chromosomes Proc Natl Acad Sci USA 98:171-175. 29. Bailey, SM, EH Goodwin, J Meyne, and M.N Cornforth 1996 CO-FISH reveals inversions associated with isochromosome formation Mutagenesis 11:139-144 30. Bailey, SM, J Meyne, MN Cornforth, TS McConnell, and E.H Goodwin 1996 A new method for detecting pericentric inversions using COD-FISH. Cytogenet Cell Genet 75:248-253. 31. Bailey, SM, EH Goodwin, and MN Cornforth. 2004 Strand-specific fluorescence in situ hybridization: the CO-FISH family. Cytogenet. Genome Res 107:14-17 32. Tanke, HJ, J Wiegant, RP van Gijlswijk, V. Bezrookove, H Pattenier, R.J Heetebrij, EG Talman, AK Raap, and J. Vrolijk 1999 New strategy for multi-colour fluorescence in situ hybridisation: COBRA: combined binary ratio labelling. Eur J Hum Genet 7:2-11 33. Wiegant, J, V Bezrookove, C Rosenberg, H.J Tanke, AK Raap, H Zhang, M Bittner, J.M Trent, and P Meltzer 2000 Differentially painting human chromosome arms with combined binary

ratio-labeling fluorescence in situ hybridization. Genome Res 10:861-865. 34. Szuhai, K, V Bezrookove, J Wiegant, J Vrolijk, R.W Dirks, C Rosenberg, AK Raap, and H.J Tanke 2000 Simultaneous molecular karyotyping and mapping of viral DNA integration sites by 25-color COBRAFISH. Genes Chromosomes Cancer 28:92-97 35. Raap, AK and HJ Tanke 2006 Combined binary ratio fluorescence in situ hybridiziation (COBRA-FISH): development and applications. Cytogenet Genome Res 114:222-226 36. Tubbs, RR, J Pettay, P Roche, MH Stoler, R. Jenkins, J Myles, and T Grogan 2000 Concomitant oncoprotein detection with fluorescence in situ hybridization (CODFISH): a fluorescence-based assay enabling simultaneous visualization of gene amplification and encoded protein expression. J Mol Diagn 2:78-83. 37. Tubbs, R, J Pettay, M Skacel, R Powell, M Stoler, P. Roche, and J Hainfeld 2002 Goldfacilitated in situ hybridization: a bright-field autometallographic alternative to fluorescence in situ hybridization for

detection of Her-2/neu gene amplification. Am J Pathol 160:15891595 38. Frada, M, F Not, I Probert, and C de Vargas. 2006 CaCO3 optical detection with fluorescent in situ hybridization: a new method to identify and quantify calcifying microorganisms from the oceans. J Phycol 42:11621169 39. Hausmann, M, R Winkler, G Hildenbrand, J. Finsterle, A Weisel, A Rapp, E Schmitt, S. Janz, and C Cremer 2003 COMBOFISH: specific labeling of nondenatured chromatin targets by computer-selected Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review DNA oligonucleotide probe combinations. BioTechniques. 35:564-577 40. Schwarz-Finsterle, J, S Stein, C Grossmann, E. Schmitt, H Schneider, L Trakhtenbrot, G. Rechavi, N Amariglio, et al. 2005 COMBO-FISH for focussed fluorescence labelling of gene domains: 3D-analysis of the genome architecture of abl and bcr in human blood cells. Cell Biol Int 29:1038-1046 41. Schwarz-Finsterle, J, S Stein, C Grossmann, E. Schmitt, L Trakhtenbrot, G Rechavi, N.

Amariglio, C Cremer, and M Hausmann. 2007 Comparison of triple helical COMBO-FISH and standard FISH by means of quantitative microscopic image analysis of abl/bcr positions in cell nuclei. J Biochem Biophys. Methods 70:397-406 42. McKenna, DJ, M Gallus, SR McKeown, C.S Downes, and VJ McKelvey-Martin 2003. Modification of the alkaline Comet assay to allow simultaneous evaluation of mitomycin C-induced DNA cross-link damage and repair of specific DNA sequences in RT4 cells. DNA Repair (Amst.) 2:879-890 43. Rapp, A, C Bock, H Dittmar, and KO Greulich. 2000 UV-A breakage sensitivity of human chromosomes as measured by COMETFISH depends on gene density and not on the chromosome size. J Photochem Photobiol B 56:109-117. 44. McKenna, DJ, NF Rajab, SR McKeown, G. McKerr, and VJ McKelvey-Martin 2003. Use of the comet-FISH assay to demonstrate repair of the TP53 gene region in two human bladder carcinoma cell lines Radiat Res 159:49-56. 45. Kumaravel, TS and RG Bristow 2005 Detection of genetic

instability at HER-2/neu and p53 loci in breast cancer cells sing CometFISH. Breast Cancer Res Treat 91:89-93 46. Amendola, R, E Basso, PG Pacifici, E Piras, A. Giovanetti, C Volpato, and G Romeo. 2006 Ret, Abl1 (cAbl) and Trp53 gene fragmentations in comet-FISH assay act as in vivo biomarkers of radiation exposure in C57BL/6 and CBA/J mice. Radiat Res 165:553-561. 47. Glei, M, A Schaeferhenrich, U Claussen, A. Kuechler, T Liehr, A Weise, B Marian, W. Sendt, and BL Pool-Zobel 2007 Comet fluorescence in situ hybridization analysis for oxidative stress-induced DNA damage in colon cancer relevant genes. Toxicol. Sci 96:279-284 48. Arutyunyan, R, A Rapp, KO Greulich, G. Hovhannisyan, S Haroutiunian, and E Gebhart. 2005 Fragility of telomeres after bleomycin and cisplatin combined treatment measured in human leukocytes with the CometFISH technique. Exp Oncol 27:38-42 49. Rapp, A, M Hausmann, and KO Greulich 2005. The comet-FISH technique: a tool for detection of specific DNA damage and

repair. Methods Mol. Biol 291:107-119 50. Branco, MR and A Pombo 2006 Intermingling of chromosome territories in interphase suggests role in translocations and transcription-dependent associations. PLoS Biol. 4:e138 51. Simonis, M, P Klous, E Splinter, Y Moshkin, R. Willemsen, E de Wit, B van Steensel, and W. de Laat 2006 Nuclear or- 402 ı BioTechniques ı www.biotechniquescom ganization of active and inactive chromatin domains uncovered by chromosome conformation capture-on-chip (4C). Nat Genet 38:1348-1354. 52. Dewald, GW, WA Wyatt, AL Juneau, R.O Carlson, AR Zinsmeister, SM Jalal, J.L Spurbeck, and RT Silver 1998 Highly sensitive fluorescence in situ hybridization method to detect double BCR/ABL fusion and monitor response to therapy in chronic myeloid leukemia. Blood 91:3357-3365 53. Grand, FH, A Chase, S Iqbal, DX Nguyen, J.L Lewis, SB Marley, RJ Davidson, JM Goldman, and M.Y Gordon 1998 A twocolor BCR-ABL probe that greatly reduces the false positive and false negative rates

for fluorescence in situ hybridization in chronic myeloid leukemia. Genes Chromosomes Cancer 23:109-115. 54. Varella-Garcia, M, CJ Hogan, LF Odom, J.L Murata-Collins, H Ai, L Chen, K Richkind, G. Paskulin, et al 2001 Minimal residual disease (MRD) in remission t(8;21) AML and in vivo differentiation detected by FISH and CD34+ cell sorting. Leukemia 15:1408-1414. 55. Brockman, SR, SF Paternoster, RP Ketterling, and G.W Dewald 2003 New highly sensitive fluorescence in situ hybridization method to detect PML/RARA fusion in acute promyelocytic leukemia. Cancer Genet Cytogenet. 145:144-151 56. Wan, TS, CC So, KC Hui, SF Yip, ES Ma, and L.C Chan 2007 Diagnostic utility of dual fusion PML/RARalpha translocation DNA probe (D-FISH) in acute promyelocytic leukemia. Oncol Rep 17:799-805 57. Shearer, BM, RA Knudson, HC Flynn, and R.P Ketterling 2005 Development of a D-FISH method to detect DEK/CAN fusion resulting from t(6;9)(p23;q34) in patients with acute myelogenous leukemia. Leukemia

19:126-131. 58. Shearer, BM, HC Flynn, RA Knudson, and R.P Ketterling 2005 Interphase FISH to detect PBX1/E2A fusion resulting from the der(19)t(1;19)(q23;p13.3) or t(1;19)(q23;p133) in paediatric patients with acute lymphoblastic leukaemia. Br J Haematol 129:45-52 59. Fernandez, JL and J Gosalvez 2002 Application of FISH to detect DNA damage. DNA breakage detection-FISH (DBD-FISH). Methods Mol. Biol 203:203-216 60. Fernandez, JL, VJ Goyanes, J Ramiro-Diaz, and J. Gosalvez 1998 Application of FISH for in situ detection and quantification of DNA breakage. Cytogenet. Cell Genet 82:251-256 61. Fernandez, JL, F Vazquez-Gundin, MT Rivero, A. Genesca, J Gosalvez, and V Goyanes. 2001 DBD-fish on neutral comets: simultaneous analysis of DNA single- and double-strand breaks in individual cells. Exp Cell Res. 270:102-109 62. Fernandez, JL, F Vazquez-Gundin, MT Rivero, V. Goyanes, and J Gosalvez 2001 Evidence of abundant constitutive alkali-labile sites in human 5 bp classical satellite DNA loci

by DBD-FISH. Mutat Res 473:163-168 63. Vazquez-Gundin, F, J Gosalvez, J de la Torre, and J.L Fernandez 2000 DNA breakage detection-fish (DBD-FISH): effect of unwinding time. Mutat Res 453:83-88 64. Enciso, M, C Lopez-Fernandez, JL Fernandez, P. Garcia, A Gosalbez, and J Gosalvez. 2006 A new method to analyze boar sperm DNA fragmentation under bright-field or fluorescence microscopy. Theriogenology 65:308-316. 65. Zhang, J, L Feuk, GE Duggan, R Khaja, and S.W Scherer 2006 Development of bioinformatics resources for display and analysis of copy number and other structural variants in the human genome. Cytogenet Genome Res 115:205-214. 66. Haaf, T and DC Ward 1994 High resolution ordering of YAC contigs using extended chromatin and chromosomes. Hum Mol Genet. 3:629-633 67. Parra, I and B Windle 1993 High resolution visual mapping of stretched DNA by fluorescent hybridization Nat Genet 5:17-21 68. Senger, G, TA Jones, H Fidlerova, P Sanseau, J. Trowsdale, M Duff, and D Sheer. 1994

Released chromatin: linearized DNA for high resolution fluorescence in situ hybridization. Hum Mol Genet 3:1275-1280 69. Florijn, RJ, LA Bonden, H Vrolijk, J Wiegant, J.W Vaandrager, F Baas, JT den Dunnen, H.J Tanke, et al 1995 Highresolution DNA fiber-FISH for genomic DNA mapping and colour bar-coding of large genes. Hum. Mol Genet 4:831-836 70. Heiskanen, M, E Hellsten, OP Kallioniemi, T.P Makela, K Alitalo, L Peltonen, and A Palotie. 1995 Visual mapping by fiber-FISH Genomics 30:31-36. 71. Bensimon, A, A Simon, A Chiffaudel, V Croquette, F. Heslot, and D Bensimon 1994 Alignment and sensitive detection of DNA by a moving interface. Science 265:2096-2098 72. Rufer, N, W Dragowska, G Thornbury, E. Roosnek, and PM Lansdorp 1998 Telomere length dynamics in human lymphocyte subpopulations measured by flow cytometry. Nat Biotechnol 16:743-747 73. Baerlocher, GM, I Vulto, G de Jong, and P.M Lansdorp 2006 Flow cytometry and FISH to measure the average length of telomeres (flow FISH). Nat

Protoc 1:2365-2376 74. Baerlocher, GM and PM Lansdorp 2003 Telomere length measurements in leukocyte subsets by automated multicolor flow-FISH. Cytometry A 55:1-6. 75. Lee, WW, KH Nam, K Terao, and Y Yoshikawa. 2002 Age-related telomere length dynamics in peripheral blood mononuclear cells of healthy cynomolgus monkeys measured by flow FISH. Immunology 105:458-465 76. Mariani, E, A Meneghetti, I Formentini, S. Neri, L Cattini, G Ravaglia, P Forti, and A. Facchini 2003 Different rates of telomere shortening and telomerase activity reduction in CD8 T and CD16 NK lymphocytes with ageing. Exp Gerontol 38:653-659 77. Ding, H, M Schertzer, X Wu, M Gertsenstein, S. Selig, M Kammori, R Pourvali, S. Poon, et al 2004 Regulation of murine telomere length by Rtel: an essential gene encoding a helicase-like protein. Cell 117:873-886. 78. Norrback, KF, M Hultdin, K Dahlenborg, P. Osterman, R Carlsson, and G Roos Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review 2001. Telomerase

regulation and telomere dynamics in germinal centers Eur J Haematol 67:309-317. 79. Alter, BP, GM Baerlocher, SA Savage, S.J Chanock, BB Weksler, JP Willner, J.A Peters, N Giri, and PM Lansdorp 2007. Very short telomere length by flow fluorescence in situ hybridization identifies patients with dyskeratosis congenita. Blood 110:1439-1447. 80. Bartolovic, K, S Balabanov, B Berner, HJ Buhring, M. Komor, S Becker, D Hoelzer, L. Kanz, et al 2005 Clonal heterogeneity in growth kinetics of CD34+CD38- human cord blood cells in vitro is correlated with gene expression pattern and telomere length. Stem Cells 23:946-957. 81. Brando, B, A Longo, B Beltrami, D Passoni, R. Verna, F Licastro, and MM Corsi. 2004 Determination of telomere length by flow-fluorescence in situ hybridization in Down’s syndrome patients. Int J Tissue React 26:61-64. 82. Brummendorf, TH, I Ersoz, U Hartmann, S. Balabanov, H Wolke, P Paschka, T Lahaye, B. Berner, et al 2003 Normalization of previously shortened telomere

length under treatment with imatinib argues against a preexisting telomere length deficit in normal hematopoietic stem cells from patients with chronic myeloid leukemia. Ann NY Acad Sci. 996:26-38 83. Derradji, H, S Bekaert, P Van Oostveldt, and S. Baatout 2005 Comparison of different protocols for telomere length estimation by combination of quantitative fluorescence in situ hybridization (Q-FISH) and flow cytometry in human cancer cell lines. Anticancer Res 25:1039-1050. 84. Scheding, S, I Ersoz, U Hartmann, K Bartolvic, S. Balabanov, A Salama, L Kanz, and T.H Brummendorf 2003 Peripheral blood cell telomere length measurements indicate that hematopoietic stem cell turnover is not significantly increased in whole blood and apheresis PLT donors. Transfusion 43:1089-1095. 85. Batliwalla, FM, N Rufer, PM Lansdorp, and P.K Gregersen 2000 Oligoclonal expansions in the CD8(+)CD28(-) T cells largely explain the shorter telomeres detected in this subset: analysis by flow FISH. Hum Immunol

61:951-958. 86. Brummendorf, TH and S Balabanov 2006. Telomere length dynamics in normal hematopoiesis and in disease states characterized by increased stem cell turnover. Leukemia 20:1706-1716. 87. Brummendorf, TH, N Rufer, TL Holyoake, J. Maciejewski, MJ Barnett, C.J Eaves, AC Eaves, N Young, and PM Lansdorp. 2001 Telomere length dynamics in normal individuals and in patients with hematopoietic stem cell-associated disorders. Ann. NY Acad Sci 938:293-303 [discussion 303-304] 88. Van Ziffle, JA, GM Baerlocher, and PM Lansdorp. 2003 Telomere length in subpopulations of human hematopoietic cells Stem Cells 21:654-660. 89. Friedrich, U and J Lenke 2006 Improved enumeration of lactic acid bacteria in meso- 404 ı BioTechniques ı www.biotechniquescom philic dairy starter cultures by using multiplex quantitative real-time PCR and flow cytometry-fluorescence in situ hybridization. Appl Environ. Microbiol 72:4163-4171 90. Jen, CJ, CH Chou, PC Hsu, SJ Yu, W.E Chen, JJ Lay, CC Huang, and FS

Wen. 2007 Flow-FISH analysis and isolation of clostridial strains in an anaerobic semi-solid bio-hydrogen producing system by hydrogenase gene target. Appl Microbiol Biotechnol 74:1126-1134. 91. Amiel, A, S Yarkoni, S Slavin, R Or, H Lorberboum-Galski, M. Fejgin, and A Nagler. 1994 Detection of minimal residual disease state in chronic myelogenous leukemia patients using fluorescence in situ hybridization. Cancer Genet Cytogenet 76:59-64 92. Di Bona, E, A Montaldi, N Guercini, V. Rossi, A Luciano, A Biondi, and F Rodeghiero. 1996 A (15;17) translocation not associated with acute promyelocytic leukaemia. Br J Haematol 95:706-709 93. Giles, RH, JG Dauwerse, C Higgins, F Petrij, J.W Wessels, GC Beverstock, H Dohner, M. Jotterand-Bellomo, et al 1997 Detection of CBP rearrangements in acute myelogenous leukemia with t(8;16). Leukemia 11:2087-2096. 94. Sacchi, N, I Magnani, L Kearney, J Wijsman, A. Hagemeijer, and M Darfler 1995. Interphase cytogenetics of the t(8;21) (q22;q22) associated

with acute myelogenous leukemia by two-color fluorescence in situ hybridization. Cancer Genet Cytogenet 79:97-103. 95. Taniwaki, M, Y Ueda, K Nishida, T Takashima, K. Kashima, F Matsuda, and G.A Silverman 1997 Rapid detection of lymphoma-specific translocations in interphase nuclei of non-Hodgkin’s lymphoma by fluorescence in situ hybridization. Leukemia 11(Suppl 3):291-293. 96. Ueda, Y, K Nishida, T Miki, S Horiike, H Kaneko, S. Yokota, S Misawa, T Abe, et al 1997. Interphase detection of BCL6/IgH fusion gene in non-Hodgkin lymphoma by fluorescence in situ hybridization Cancer Genet Cytogenet. 99:102-107 97. Li, JY, F Gaillard, A Moreau, JL Harousseau, C. Laboisse, N Milpied, R. Bataille, and H Avet-Loiseau 1999 Detection of translocation t(11;14)(q13;q32) in mantle cell lymphoma by fluorescence in situ hybridization. Am J Pathol 154:1449-1452 98. Ma, SK, TS Wan, AT Cheuk, LF Fung, G.C Chan, SY Chan, SY Ha, and LC Chan. 2001 Characterization of additional genetic events in childhood

acute lymphoblastic leukemia with TEL/AML1 gene fusion: a molecular cytogenetics study. Leukemia 15:1442-1447 99. Martineau, M, GR Jalali, KE Barber, ZJ Broadfield, K.L Cheung, J Lilleyman, AV Moorman, S. Richards, et al 2005 ETV6/ RUNX1 fusion at diagnosis and relapse: some prognostic indications. Genes Chromosomes Cancer 43:54-71. 100. Martinez-Ramirez, A, M Urioste, T Contra, A. Cantalejo, A Tavares, JA Portero, B. Lopez-Ibor, M Bernacer, et al 2001 Fluorescence in situ hybridization study of TEL/ AML1 fusion and other abnormalities involving TEL and AML1 genes. Correlation with cytoge- netic findings and prognostic value in children with acute lymphocytic leukemia. Haematologica 86:1245-1253. 101. Konig, M, M Reichel, R Marschalek, OA Haas, and S. Strehl 2002 A highly specific and sensitive fluorescence in situ hybridization assay for the detection of t(4;11)(q21;q23) and concurrent submicroscopic deletions in acute leukaemias. Br J Haematol 116:758-764 102. Chase, A, F Grand, JG

Zhang, N Blackett, J. Goldman, and M Gordon 1997 Factors influencing the false positive and negative rates of BCR-ABL fluorescence in situ hybridization. Genes Chromosomes Cancer 18:246-253. 103. Chen, Z, M Notohamiprodjo, PD Richards, F.B Lane, R Morgan, JF Stone, and A.A Sandberg 1997 Some observations on fluorescence in situ hybridization evaluation of chronic myelocytic leukemia. Cancer Genet Cytogenet. 98:1-3 104. Hsu, WT, H Preisler, K Szego, R Sprudzs, and X.Z Gao 1998 The ABL/BCR fusion gene on chromosome 9 in Ph-negative chronic myelogenous leukemia: a case for vigilance in fluorescence in situ hybridization interpretation. Cancer Genet. Cytogenet 104:57-60 105. Gerdes, MG, KC Carter, PT Moen, Jr, and J.B Lawrence 1994 Dynamic changes in the higher-level chromatin organization of specific sequences revealed by in situ hybridization to nuclear halos. J Cell Biol 126:289-304 106. Wiegant, J, W Kalle, L Mullenders, S Brookes, J.M Hoovers, JG Dauwerse, GJ van Ommen, and A.K Raap

1992 Highresolution in situ hybridization using DNA halo preparations. Hum Mol Genet 1:587-591 107. Luderus, ME, B van Steensel, L Chong, O.C Sibon, FF Cremers, and T de Lange 1996. Structure, subnuclear distribution, and nuclear matrix association of the mammalian telomeric complex. J Cell Biol 135:867-881 108. Heng, HH, S Goetze, CJ Ye, G Liu, JB Stevens, S.W Bremer, SM Wykes, J Bode, and S.A Krawetz 2004 Chromatin loops are selectively anchored using scaffold/matrix-attachment regions. J Cell Sci 117:999-1008 109. Iarovaia, OV, SB Akopov, LG Nikolaev, E.D Sverdlov, and SV Razin 2005 Induction of transcription within chromosomal DNA loops flanked by MAR elements causes an association of loop DNA with the nuclear matrix. Nucleic Acids Res. 33:4157-4163 110. Iarovaia, OV, A Bystritskiy, D Ravcheev, R Hancock, and S.V Razin 2004 Visualization of individual DNA loops and a map of loop domains in the human dystrophin gene. Nucleic Acids Res. 32:2079-2086 111. Ratsch, A, S Joos, P

Kioschis, and P Lichter. 2002 Topological organization of the MYC/IGK locus in Burkitt’s lymphoma cells assessed by nuclear halo preparations. Exp Cell Res. 273:12-20 112. Croft, JA, JM Bridger, S Boyle, P Perry, P Teague, and W.A Bickmore 1999 Differences in the localization and morphology of chromosomes in the human nucleus. J Cell Biol 145:1119-1131. 113. Ma, H, AJ Siegel, and R Berezney 1999 Association of chromosome territories with the nuclear matrix. Disruption of human chromosome territories correlates with the Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review release of a subset of nuclear matrix proteins. J Cell Biol 146:531-542 114. Raap, AK and J Wiegant 1994 Use of DNAhalo preparations for high-resolution DNA in situ hybridization. Methods Mol Biol 33:123-130 115. Repping, S, JW de Vries, SK van Daalen, C.M Korver, NJ Leschot, and F van der Veen. 2003 The use of spermHALO-FISH to determine DAZ gene copy number. Mol Hum Reprod. 9:183-188 116. Jordan, R, J

Edington, HH Evans, and J.L Schwartz 1999 Detection of chromosome aberrations by FISH as a function of cell division cycle (harlequin-FISH). BioTechniques 26:532-534. 117. Kulka, U, R Huber, P Muller, S Knehr, and M. Bauchinger 1995 Combined FISH painting and harlequin staining for cell cycle-controlled chromosome analysis in human lymphocytes. Int. J Radiat Biol 68:25-27 118. Latt, SA 1973 Microfluorometric detection of deoxyribonucleic acid replication in human metaphase chromosomes. Proc Natl Acad Sci USA 70:3395-3399. 119. Perry, P and S Wolff 1974 New Giemsa method for the differential staining of sister chromatids. Nature 251:156-158 120. Sullivan, GJ, JM Bridger, AP Cuthbert, R.F Newbold, WA Bickmore, and B McStay. 2001 Human acrocentric chromosomes with transcriptionally silent nucleolar organizer regions associate with nucleoli. EMBO J. 20:2867-2874 121. Shiels, C, SA Islam, R Vatcheva, P Sasieni, M.J Sternberg, PS Freemont, and D Sheer 2001. PML bodies associate specifically

with the MHC gene cluster in interphase nuclei. J Cell Sci. 114:3705-3716 122. Bridger, JM, IR Kill, and P Lichter 1998 Association of pKi-67 with satellite DNA of the human genome in early G1 cells. Chromosome Res. 6:13-24 123. Brown, KE, SS Guest, ST Smale, K Hahm, M. Merkenschlager, and AG Fisher 1997 Association of transcriptionally silent genes with Ikaros complexes at centromeric heterochromatin. Cell 91:845-854 124. Zirbel, RM, UR Mathieu, A Kurz, T Cremer, and P. Lichter 1993 Evidence for a nuclear compartment of transcription and splicing located at chromosome domain boundaries. Chromosome Res. 1:93-106 125. Page, SL, WC Earnshaw, KH Choo, and L.G Shaffer 1995 Further evidence that CENP-C is a necessary component of active centromeres: studies of a dic(X;15) with simultaneous immunofluorescence and FISH. Hum Mol Genet. 4:289-294 126. Zinner, R, K Teller, R Versteeg, T Cremer, and M. Cremer 2007 Biochemistry meets nuclear architecture: multicolor immuno-FISH for co-localization

analysis of chromosome segments and differentially expressed gene loci with various histone methylations. Adv Enzyme Regul. 47:223-241 127. Koshkin, AA and J Wengel 1998 Synthesis of novel 2′,3′-linked bicyclic thymine ribonucleosides. J Org Chem 63:2778-2781 128. Petersen, M, JT Nielsen, K Bondensgaard, J. Wengel, and JP Jacobsen 2003 Structural characterization of LNA and alpha-L-LNA 406 ı BioTechniques ı www.biotechniquescom hybridized to RNA. Nucleosides Nucleotides Nucleic Acids 22:1691-1693. 129. Silahtaroglu, A, H Pfundheller, A Koshkin, N. Tommerup, and S Kauppinen 2004. LNA-modified oligonucleotides are highly efficient as FISH probes. Cytogenet Genome Res. 107:32-37 130. Silahtaroglu, AN, N Tommerup, and H Vissing. 2003 FISHing with locked nucleic acids (LNA): evaluation of different LNA/DNA mixmers. Mol Cell Probes 17:165-169 131. Speicher, MR, S Gwyn Ballard, and DC Ward. 1996 Karyotyping human chromosomes by combinatorial multi-fluor FISH. Nat Genet 12:368-375.

132. Nietzel, A, M Rocchi, H Starke, A Heller, W. Fiedler, I Wlodarska, IF Loncarevic, V Beensen, et al. 2001 A new multicolor-FISH approach for the characterization of marker chromosomes: centromere-specific multicolor-FISH (cenM-FISH). Hum Genet 108:199-204 133. Henegariu, O, P Bray-Ward, S Artan, G.H Vance, M Qumsyieh, and DC Ward 2001. Small marker chromosome identification in metaphase and interphase using centromeric multiplex fish (CM-FISH) Lab Invest. 81:475-481 134. Starke, H, A Nietzel, A Weise, A Heller, K Mrasek, B. Belitz, C Kelbova, M Volleth, et al. 2003 Small supernumerary marker chromosomes (SMCs): genotype-phenotype correlation and classification. Hum Genet 114:51-67 135. Brown, J, K Saracoglu, S Uhrig, MR Speicher, R. Eils, and L Kearney 2001 Subtelomeric chromosome rearrangements are detected using an innovative 12-color FISH assay (M-TEL). Nat Med 7:497-501 136. Fauth, C, H Zhang, S Harabacz, J Brown, K. Saracoglu, G Lederer, O Rittinger, I Rost, et al. 2001 A new

strategy for the detection of subtelomeric rearrangements. Hum Genet 109:576-583. 137. Schrock, E, S du Manoir, T Veldman, B Schoell, J. Wienberg, MA Ferguson-Smith, Y. Ning, DH Ledbetter, et al 1996 Multicolor spectral karyotyping of human chromosomes. Science 273:494-497. 138. Kearney, L 2006 Multiplex-FISH (M-FISH): technique, developments and applications. Cytogenet. Genome Res 114:189-198 139. Schrock, E, P Zschieschang, P O’Brien, A. Helmrich, T Hardt, A Matthaei, and K Stout-Weider. 2006 Spectral karyotyping of human, mouse, rat and ape chromosomes applications for genetic diagnostics and research. Cytogenet. Genome Res 114:199-221 140. Ligon, AH, AL Beaudet, and LG Shaffer 1997. Simultaneous, multilocus FISH analysis for detection of microdeletions in the diagnostic evaluation of developmental delay and mental retardation. Am J Hum Genet 61:51-59 141. Eckel, H, J Kleinstein, P Wieacker, and M Stumm. 2003 Multi-locus (ML)-FISH is a reliable tool for nondisjunction studies in

human oocytes Cytogenet Genome Res 103:47-53 142. Brown, JM, J Evans, and MS Kovacs 1992 The prediction of human tumor radiosensitivity in situ: an approach using chromosome aberrations detected by fluorescence in situ hybridization. Int. J Radiat Oncol Biol Phys 24:279-286 143. Coco-Martin, JM and AC Begg 1997 Detection of radiation-induced chromosome ab- errations using fluorescence in situ hybridization in drug-induced premature chromosome condensations of tumour cell lines with different radiosensitivities. Int J Radiat Biol 71:265-273 144. Evans, JW, JA Chang, AJ Giaccia, D Pinkel, and J.M Brown 1991 The use of fluorescence in situ hybridisation combined with premature chromosome condensation for the identification of chromosome damage. Br J Cancer 63:517-521. 145. Lee, R, S Yamada, N Yamamoto, T Miyamoto, K. Ando, M Durante, and H Tsujii. 2004 Chromosomal aberrations in lymphocytes of lung cancer patients treated with carbon ions J Radiat Res (Tokyo) 45:195-199 146. M’Kacher,

R, D Violot, B Aubert, T Girinsky, J. Dossou, N Beron-Gaillard, P Carde, and C. Parmentier 2003 Premature chromosome condensation associated with fluorescence in situ hybridisation detects cytogenetic abnormalities after a CT scan: evaluaton of the low-dose effect. Radiat Prot. Dosimetry 103:35-40 147. Pouget, JP, C Laurent, M Delbos, M Benderitter, I. Clairand, F Trompier, J Stephanazzi, H. Carsin, et al 2004 PCC-FISH in skin fibroblasts for local dose assessment: biodosimetric analysis of a victim of the Georgian radiological accident. Radiat Res 162:365-376 148. Bergs, JW, NA Franken, R ten Cate, C van Bree, and J. Haveman 2006 Effects of cisplatin and gamma-irradiation on cell survival, the induction of chromosomal aberrations and apoptosis in SW-1573 cells. Mutat Res 594:148-154 149. Franken, NA, JW Bergs, TT Kok, RR Kuperus, F. Stecher-Rasmussen, J Haveman, C. Van Bree, and LJ Stalpers 2006 Gadolinium enhances the sensitivity of SW-1573 cells for thermal neutron irradiation.

Oncol Rep 15:715-720. 150. Nielsen, PE, M Egholm, RH Berg, and O Buchardt. 1991 Sequence-selective recognition of DNA by strand displacement with a thyminesubstituted polyamide. Science 254:1497-1500 151. Lansdorp, PM, NP Verwoerd, FM van de Rijke, V. Dragowska, MT Little, RW Dirks, A.K Raap, and HJ Tanke 1996 Heterogeneity in telomere length of human chromosomes. Hum Mol Genet 5:685-691 152. de Pauw, ES, NP Verwoerd, N Duinkerken, R. Willemze, AK Raap, WE Fibbe, and HJ Tanke. 1998 Assessment of telomere length in hematopoietic interphase cells using in situ hybridization and digital fluorescence microscopy. Cytometry 32:163-169. 153. Giesen, U, W Kleider, C Berding, A Geiger, H. Orum, and PE Nielsen 1998 A formula for thermal stability (Tm) prediction of PNA/DNA duplexes. Nucleic Acids Res 26:5004-5006 154. Chen, C, YK Hong, SD Ontiveros, M Egholm, and W.M Strauss 1999 Single base discrimination of CENP-B repeats on mouse and human chromosomes with PNA-FISH. Mamm Genome 10:13-18. 155.

Chen, C, B Wu, T Wei, M Egholm, and W.M Strauss 2000 Unique chromosome identification and sequence-specific structural analysis with short PNA oligomers Mamm Genome 11:384-391. 156. Taneja, KL, EA Chavez, J Coull, and PM Lansdorp. 2001 Multicolor fluorescence in situ hybridization with peptide nucleic acid probes for Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review enumeration of specific chromosomes in human cells. Genes Chromosomes Cancer 30:57-63 157. Pellestor, F, B Andreo, K Taneja, and B Williams. 2003 PNA on human sperm: a new approach for in situ aneuploidy estimation. Eur J. Hum Genet 11:337-341 158. Paulasova, P, B Andreo, J Diblik, M Macek, and F. Pellestor 2004 The peptide nucleic acids as probes for chromosomal analysis: application to human oocytes, polar bodies and preimplantation embryos. Mol Hum Reprod 10:467-472 159. Larsen, RD, A Schonau, M Thisted, KH Petersen, J. Lohse, B Christensen, J Philip, and K.J Pluzek 2003 Detection of gammaglobin mRNA in

fetal nucleated red blood cells by PNA fluorescence in situ hybridization. Prenat. Diagn 23:52-59 160. Lefmann, M, B Schweickert, P Buchholz, U.B Gobel, T Ulrichs, P Seiler, D Theegarten, and A. Moter 2006 Evaluation of peptide nucleic acid-fluorescence in situ hybridization for identification of clinically relevant mycobacteria in clinical specimens and tissue sections. J Clin Microbiol. 44:3760-3767 161. Stender, H 2003 PNA FISH: an intelligent stain for rapid diagnosis of infectious diseases. Expert Rev. Mol Diagn 3:649-655 162. Martens, UM, JM Zijlmans, SS Poon, W Dragowska, J. Yui, EA Chavez, RK Ward, and P.M Lansdorp 1998 Short telomeres on human chromosome 17p. Nat Genet 18:76-80 163. Hultdin, M, E Gronlund, K Norrback, E Eriksson-Lindstrom, T. Just, and G Roos 1998. Telomere analysis by fluorescence in situ hybridization and flow cytometry. Nucleic Acids Res. 26:3651-3656 164. Poon, SS, UM Martens, RK Ward, and P.M Lansdorp 1999 Telomere length measurements using digital

fluorescence microscopy Cytometry 36:267-278. 165. Slijepcevic, P 2001 Telomere length measurement by Q-FISH Methods Cell Sci 23:17-22 166. Baird, DM, J Rowson, D Wynford-Thomas, and D. Kipling 2003 Extensive allelic variation and ultrashort telomeres in senescent human cells. Nat Genet 33:203-207 167. Nagele, RG, AQ Velasco, WJ Anderson, D.J McMahon, Z Thomson, J Fazekas, K Wind, and H. Lee 2001 Telomere associations in interphase nuclei: possible role in maintenance of interphase chromosome topology. J Cell Sci 114:377-388. 168. Narath, R, T Lorch, KM Greulich-Bode, P Boukamp, and P.F Ambros 2005 Automatic telomere length measurements in interphase nuclei by IQ-FISH. Cytometry A 68:113-120 169. Xiao, Y and PE Barker 2004 Semiconductor nanocrystal probes for human metaphase chromosomes. Nucleic Acids Res 32:e28 170. Pathak, S, SK Choi, N Arnheim, and ME Thompson. 2001 Hydroxylated quantum dots as luminescent probes for in situ hybridization. J Am. Chem Soc 123:4103-4104 171. Wu, SM, X

Zhao, ZL Zhang, HY Xie, Z.Q Tian, J Peng, ZX Lu, DW Pang, and Z.X Xie 2006 Quantum-dot-labeled DNA probes for fluorescence in situ hybridization (FISH) in the microorganism Escherichia coli. Chemphyschem 7:1062-1067. 172. Chan, P, T Yuen, F Ruf, J Gonzalez-Maeso, and S.C Sealfon 2005 Method for multiplex cellular detection of mRNAs using quantum dot 408 ı BioTechniques ı www.biotechniquescom fluorescent in situ hybridization. Nucleic Acids Res. 33:e161 173. Bentolila, LA and S Weiss 2006 Single-step multicolor fluorescence in situ hybridization using semiconductor quantum dot-DNA conjugates. Cell Biochem Biophys 45:59-70 174. Sunamura, M and A Maruyama 2006 A digital imaging procedure for seven-probelabeling FISH (Rainbow-FISH) and its application to estuarine microbial communities. FEMS Microbiol. Ecol 55:159-166 175. Takada, T, K Matsumoto, and K Nomoto 2004. Development of multi-color FISH method for analysis of seven Bifidobacterium species in human feces. J Microbiol Methods

58:413-421 176. Loy, A, M Horn, and M Wagner 2003 probeBase: an online resource for rRNA-targeted oligonucleotide probes. Nucleic Acids Res 31:514-516. 177. Maruyama, A and M Sunamura 2000 Simultaneous direct counting of total and specific microbial cells in seawater, using a deepsea microbe as target. Appl Environ Microbiol 66:2211-2215. 178. Huang, WE, K Stoecker, R Griffiths, L Newbold, H. Daims, AS Whiteley, and M Wagner. 2007 Raman-FISH: combining stableisotope Raman spectroscopy and fluorescence in situ hybridization for the single cell analysis of identity and function. Environ Microbiol 9:1878-1889. 179. Zou, Y, SM Gryaznov, JW Shay, WE Wright, and M.N Cornforth 2004 Asynchronous replication timing of telomeres at opposite arms of mammalian chromosomes. Proc. Natl Acad Sci USA 101:12928-12933 180. Lichter, P, SA Ledbetter, DH Ledbetter, and D.C Ward 1990 Fluorescence in situ hybridization with Alu and L1 polymerase chain reaction probes for rapid characterization of human

chromosomes in hybrid cell lines. Proc Natl Acad. Sci USA 87:6634-6638 181. Carter, NP, MA Ferguson-Smith, MT Perryman, H. Telenius, AH Pelmear, MA Leversha, M.T Glancy, SL Wood, et al 1992 Reverse chromosome painting: a method for the rapid analysis of aberrant chromosomes in clinical cytogenetics. J Med Genet 29:299-307 182. de Pater, J, C Van der Sijs-Bos, M Prins, J. Derks, J Albrechts, and J Engelen 2006 Prenatal identification of a marker chromosome 16 by chromosome microdissection and reverse FISH. Eur J Med Genet 49:306-312 183. Li, YC, YM Cheng, LJ Hsieh, OA Ryder, F. Yang, SJ Liao, KM Hsiao, FJ Tsai, et al. 2005 Karyotypic evolution of a novel cervid satellite DNA family isolated by microdissection from the Indian muntjac Y-chromosome. Chromosoma 114:28-38. 184. Blennow, E, H Telenius, C Larsson, D de Vos, S. Bajalica, BA Ponder, and M Nordenskjold. 1992 Complete characterization of a large marker chromosome by reverse and forward chromosome painting. Hum Genet 90:371-374.

185. Kozma, C, J Blancato, J Meck, and Y Jiang 1998. Characterization of a supernumerary marker derived from chromosome 17 by microdissection in an adult with MR/MCA Am J Med Genet. 77:19-22 186. Mahjoubi, F, GB Peters, P Malafiej, C Shalhoub, A. Turner, A Daniel, and RJ Hill. 2005 An analphoid marker chromosome inv dup(15)(q26.1qter), detected during prenatal diagnosis and characterized via chromosome microdissection. Cytogenet Genome Res 109:485490 187. Mahjoubi, F, RJ Hill, and GB Peters 2006 Chromosome microdissection identifies genomic amplifications associated with drug resistance in a leukemia cell line: an approach to understanding drug resistance in cancer. Chromosome Res 14:263-276. 188. Zwirglmaier, K, K Fichtl, and W Ludwig 2005. In situ functional gene analysis: recognition of individual genes by fluorescence in situ hybridization. Methods Enzymol 397:338-351 189. Zwirglmaier, K, W Ludwig, and KH Schleifer. 2004 Recognition of individual genes in a single bacterial cell

by fluorescence in situ hybridizationRING-FISH. Mol Microbiol 51:89-96. 190. Femino, AM, FS Fay, K Fogarty, and RH Singer. 1998 Visualization of single RNA transcripts in situ Science 280:585-590 191. Narimatsu, R and BK Patterson 2005 High-throughput cervical cancer screening using intracellular human papillomavirus E6 and E7 mRNA quantification by flow cytometry. Am J Clin. Pathol 123:716-723 192. Popescu, NC, MC Chen, S Simpson, S Solinas, and J.A DiPaolo 1993 A Burkitt lymphoma cell line with integrated Epstein-Barr virus at a stable chromosome modification site. Virology 195:248-251. 193. Fan, Y, SA Braut, Q Lin, RH Singer, and A.I Skoultchi 2001 Determination of transgenic loci by expression FISH Genomics 71:66-69 194. Braidotti, G 2001 RNA-FISH to analyze allele-specific expression. Methods Mol Biol 181:169-180. 195. Herzing, LB, EH Cook, Jr, and DH Ledbetter. 2002 Allele-specific expression analysis by RNA-FISH demonstrates preferential maternal expression of UBE3A and imprint

maintenance within 15q11-q13 duplications. Hum. Mol Genet 11:1707-1718 196. Levsky, JM, SM Shenoy, RC Pezo, and R.H Singer 2002 Single-cell gene expression profiling. Science 297:836-840 197. Bridger, JM, C Kalla, H Wodrich, S Weitz, J.A King, K Khazaie, HG Krausslich, and P. Lichter 2005 Nuclear RNAs confined to a reticular compartment between chromosome territories. Exp Cell Res 302:180-193 198. Clemson, CM, JA McNeil, HF Willard, and J.B Lawrence 1996 XIST RNA paints the inactive X chromosome at interphase: evidence for a novel RNA involved in nuclear/chromosome structure. J Cell Biol 132:259-275 199. Dirks, RW and AK Raap 1995 Cell-cycledependent gene expression studied by two-colour fluorescent detection of a mRNA and histone mRNA. Histochem Cell Biol 104:391-395 200. Gribnau, J, K Diderich, S Pruzina, R Calzolari, and P. Fraser 2000 Intergenic transcription and developmental remodeling of chromatin subdomains in the human beta-globin locus. Mol Cell 5:377-386 201. Hutchinson, JN,

AW Ensminger, CM Clemson, C.R Lynch, JB Lawrence, and A Chess. 2007 A screen for nuclear transcripts identifies two linked noncoding RNAs associated Vol. 45 ı No 4 ı 2008 Source: http://www.doksinet Review with SC35 splicing domains. BMC Genomics 8:39. 202. Lampel, S, JM Bridger, RM Zirbel, UR Mathieu, and P. Lichter 1997 Nuclear RNA accumulations contain released transcripts and exhibit specific distributions with respect to Sm antigen foci. DNA Cell Biol 16:1133-1142 203. Lawrence, JB, RH Singer, and LM Marselle. 1989 Highly localized tracks of specific transcripts within interphase nuclei visualized by in situ hybridization Cell 57:493-502 204. Miles, J, JA Mitchell, L Chakalova, B Goyenechea, C.S Osborne, L O’Neill, K Tanimoto, J.D Engel, and P Fraser 2007 Intergenic transcription, cell-cycle and the developmentally regulated epigenetic profile of the human beta-globin locus. PLoS One 2:e630 205. van de Corput, MP and FG Grosveld 2001 Fluorescence in situ hybridization

analysis of transcript dynamics in cells. Methods 25:111118 206. Bonifazi, E, F Gullotta, L Vallo, R Iraci, A.M Nardone, E Brunetti, A Botta, and G Novelli. 2006 Use of RNA fluorescence in situ hybridization in the prenatal molecular diagnosis of myotonic dystrophy type I. Clin Chem 52:319-322. 207. Muller, S and J Wienberg 2001 “Bar-coding” primate chromosomes: molecular cytogenetic screening for the ancestral hominoid karyotype. Hum. Genet 109:85-94 208. Muller, S, PC O’Brien, MA FergusonSmith, and J Wienberg 1997 A novel source of highly specific chromosome painting probes for human karyotype analysis derived from primate homologues. Hum Genet 101:149-153 209. Muller, S, PC O’Brien, MA FergusonSmith, and J Wienberg 1997 Reciprocal chromosome painting between human and prosimians (Eulemur macaco macaco and E. fulvus mayottensis) Cytogenet Cell Genet 78:260-271 210. Muller, S, PC O’Brien, MA FergusonSmith, and J Wienberg 1998 Cross-species colour segmenting: a novel tool in

human karyotype analysis. Cytometry 33:445-452 211. Zhao, L, K Hayes, and A Glassman 2000 Enhanced detection of chromosomal abnormalities with the use of RxFISH multicolor banding technique. Cancer Genet Cytogenet 118:108111 212. Micci, F, MR Teixeira, CU Dietrich, G Saeter, B. Bjerkehagen, and S Heim 1999 Combined RxFISH/G-banding allows refined karyotyping of solid tumors. Hum Genet 104:370-375. 213. Teixeira, MR, F Micci, CU Dietrich, and S Heim. 1999 Cross-species color banding characterization of chromosomal rearrangements in leukemias with incomplete G-band karyotypes. Genes Chromosomes Cancer 26:13-19. 214. Teixeira, MR, F Micci, CU Dietrich, and S Heim. 2000 Detailed genome-wide screening for inter- and intrachromosomal abnormalities by sequential G-banding and RxFISH color banding of the same metaphase cells. Cancer Genet Cytogenet. 119:94-101 215. van der Burg, M, TS Poulsen, SP Hunger, H.B Beverloo, EM Smit, K Vang-Nielsen, A.W Langerak, and JJ van Dongen 2004 Split-signal

FISH for detection of chromosome aberrations in acute lymphoblastic leukemia. Leukemia 18:895-908. Vol. 45 ı No 4 ı 2008 216. Siebert, R, P Matthiesen, S Harder, Y Zhang, A. Borowski, R Zuhlke-Jenisch, S Metzke, S Joos, et al. 1998 Application of interphase fluorescence in situ hybridization for the detection of the Burkitt translocation t(8;14)(q24;q32) in B-cell lymphomas. Blood 91:984-990 217. Siebert, R, P Matthiesen, S Harder, Y Zhang, A. Borowski, R Zuhlke-Jenisch, H Plendl, S. Metzke, et al 1998 Application of interphase cytogenetics for the detection of t(11;14) (q13;q32) in mantle cell lymphomas. Ann Oncol 9:519-526. 218. Raap, AK, MP van de Corput, RA Vervenne, R.P van Gijlswijk, HJ Tanke, and J. Wiegant 1995 Ultra-sensitive FISH using peroxidase-mediated deposition of biotinor fluorochrome tyramides Hum Mol Genet 4:529-534. 219. Schriml, LM, HM Padilla-Nash, A Coleman, P. Moen, WG Nash, J Menninger, G. Jones, T Ried, and M Dean 1999 Tyramide signal amplification

(TSA)-FISH applied to mapping PCR-labeled probes less than 1 kb in size. BioTechniques 27:608-613. 220. Van Tine, BA, TR Broker, and LT Chow 2005. Simultaneous in situ detection of RNA, DNA, and protein using tyramide-coupled immunofluorescence. Methods Mol Biol 292:215230 221. Van Tine, BA, J Knops, GM Shaw, and W.A May 1999 Assignment of human MFNG, manic fringe Drosophila homolog, to 22q13.1 using tyramide fluorescence in situ hybridization (T-FISH). Cytogenet Cell Genet 87:132-133 222. Van Tine, BA, JF Knops, A Butler, P Deloukas, G.M Shaw, and PH King 1998 Localization of HuC (ELAVL3) to chromosome 19p13.2 by fluorescence in situ hybridization utilizing a novel tyramide labeling technique. Genomics 53:296-299. 223. Tibiletti, MG, B Bernasconi, A Dionigi, and C. Riva 1999 The applications of FISH in tumor pathology. Adv Clin Path 3:111-118 224. Nomura, K, T Yoshino, S Nakamura, Y Akano, H. Tagawa, K Nishida, M Seto, S Nakamura, et al. 2003 Detection of t(11;18) (q21;q21) in

marginal zone lymphoma of mucosa-associated lymphocytic tissue type on paraffinembedded tissue sections by using fluorescence in situ hybridization. Cancer Genet Cytogenet 140:49-54. 225. Erjavec-Skerget, A, S Stangler-Herodez, A. Zagorac, B Zagradisnik, and N KokaljVokac 2006 Subtelomeric chromosome rearrangements in children with idiopathic mental retardation: applicability of three molecular-cytogenetic methods. Croat Med J 47:841-850 226. Izumi, H, T Hara, A Oga, K Matsuda, Y Sato, K. Naito, and K Sasaki 2002 High telomerase activity correlates with the stabilities of genome and DNA ploidy in renal cell carcinoma. Neoplasia 4:103-111. 227. Scheel, C, KL Schaefer, A Jauch, M Keller, D. Wai, C Brinkschmidt, F van Valen, W Boecker, et al. 2001 Alternative lengthening of telomeres is associated with chromosomal instability in osteosarcomas. Oncogene 20:38353844 228. Cremer, T, A Kurz, R Zirbel, S Dietzel, B Rinke, E. Schrock, MR Speicher, U Mathieu, et al. 1993 Role of chromosome

territories in the functional compartmentalization of the cell nucleus. Cold Spring Harb Symp Quant Biol 58:777-792. 229. Kurz, A, S Lampel, JE Nickolenko, J Bradl, A. Benner, RM Zirbel, T Cremer, and P Lichter. 1996 Active and inactive genes localize preferentially in the periphery of chromosome territories. J Cell Biol 135:1195-1205 230. Hepperger, C, S Otten, J von Hase, and S. Dietzel 2007 Preservation of large-scale chromatin structure in FISH experiments. Chromosoma 116:117-133. 231. Solovei, I, A Cavallo, L Schermelleh, F Jaunin, C. Scasselati, D Cmarko, C Cremer, S. Fakan, and T Cremer 2002 Spatial preservation of nuclear chromatin architecture during three-dimensional fluorescence in situ hybridization (3D-FISH). Exp Cell Res 276:10-23 232. Bolzer, A, G Kreth, I Solovei, D Koehler, K Saracoglu, C. Fauth, S Muller, R Eils, et al 2005. Three-dimensional maps of all chromosomes in human male fibroblast nuclei and prometaphase rosettes PLoS Biol 3:e157 233. Scherthan, H, T

Cremer, U Arnason, HU Weier, A. Lima-de-Faria, and L Fronicke 1994. Comparative chromosome painting discloses homologous segments in distantly related mammals. Nat Genet 6:342-347 234. Chowdhary, BP, T Raudsepp, L Fronicke, and H. Scherthan 1998 Emerging patterns of comparative genome organization in some mammalian species as revealed by Zoo-FISH. Genome Res. 8:577-589 235. Ferguson-Smith, MA, F Yang, W Rens, and P.C O’Brien 2005 The impact of chromosome sorting and painting on the comparative analysis of primate genomes. Cytogenet Genome Res 108:112-121. 236. Froenicke, L 2005 Origins of primate chromosomesas delineated by Zoo-FISH and alignments of human and mouse draft genome sequences. Cytogenet Genome Res 108:122138 237. Froenicke, L, MG Caldes, A Graphodatsky, S. Muller, LA Lyons, TJ Robinson, M Volleth, F. Yang, and J Wienberg 2006 Are molecular cytogenetics and bioinformatics suggesting diverging models of ancestral mammalian genomes? Genome Res. 16:306-310 Received 22

October 2007; accepted 30 January 2008. Address correspondence to Emanuela V. Volpi, Molecular Cytogenetics Laboratory, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK. e-mail: evolpi@well ox.acuk and/or Joanna M Bridger, Laboratory of Nuclear and Genomic Health, Centre for Cell and Chromosome Biology, Division of Biosciences, School of Health Sciences and Social Care, Brunel University, Uxbridge, UK. e-mail: joanna.bridger@brunelacuk To purchase reprints of this article, contact: Reprints@BioTechniques.com www.biotechniquescom ı BioTechniques ı 409