Germline, Somatic and Epigenetic Events Underlying Mismatch Repair Deficiency in Colorectal and HNPCC-Related Cancers

Total Page:16

File Type:pdf, Size:1020Kb

Germline, Somatic and Epigenetic Events Underlying Mismatch Repair Deficiency in Colorectal and HNPCC-Related Cancers Oncogene (2002) 21, 7585 – 7592 ª 2002 Nature Publishing Group All rights reserved 0950 – 9232/02 $25.00 www.nature.com/onc Germline, somatic and epigenetic events underlying mismatch repair deficiency in colorectal and HNPCC-related cancers Siu Tsan Yuen*,1,3, Tsun Leung Chan1, Judy WC Ho2,3, Annie SY Chan1, Lap Ping Chung1, Polly WY Lam4, Chun Wah Tse5, Andrew H Wyllie6 and Suet Yi Leung*,1 1Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Hong Kong; 2Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Hong Kong; 3Hereditary Gastrointestinal Cancer Registry, The University of Hong Kong, Queen Mary Hospital, Hong Kong; 4Department of Pathology, Queen Elizabeth Hospital, Hong Kong; 5Department of Surgery, Queen Elizabeth Hospital, Hong Kong; 6Department of Pathology, Cambridge University, UK High-frequency microsatellite instability (MSI-H) results wild-type RNA by as yet undiscovered mechanisms, or from deficiency in nucleotide mismatch repair. It no detectable abnormality in any of these parameters. contributes significantly to carcinogenesis in the human Evidence is presented to indicate that methylation in colorectal mucosa. Here we study 41 colorectal and three proximal region of the hMLH1 promoter is a more other HNPCC-related cancers with MSI-H to provide reliable correlate of transcriptional silencing in colorectal comprehensive information on the mechanisms of cancers than methylation in upstream region. These inactivation of the two major proteins involved, hMLH1 observations have significant implications for manage- and hMSH2. Seventeen of the patients had family ment of patients with MSI-H tumours. histories meeting the criteria for Bethesda grades 1, 2 Oncogene (2002) 21, 7585 – 7592. doi:10.1038/sj.onc. or 3. Of these familial cases, 14 (83%) had early-onset 1205968 disease, defined on the basis of diagnosis prior to the age of 50, but in three the disease was of late onset (450 Keywords: microsatellite instability; promoter methyla- years). A second subset of 20 patients had early onset tion; mismatch repair gene mutation; colorectal cancer; disease without family history. The remaining seven HNPCC patients were selected to allow comparisons with sporadic, late-onset disease, the molecular basis of which has been extensively reported elsewhere. We stratified the tumours initially on the basis of hMLH1 or hMSH2 protein deficiency, detected by immunohistochemistry, High frequency microsatellite instability (MSI-H) is the and then by analysis of germline and somatic mutation, phenotypic manifestation of mismatch repair defect. In mRNA transcription, loss of heterozygosity (LOH) at colorectal carcinomas most MSI-H is associated with the hMLH1 and hMSH2 loci, and methylation status in inactivation of either hMSH2 or hMLH1, genes that two regions of the hMLH1 promoter. The functional encode proteins involved early in the process of significance of several of these changes in the MSI-H mismatch repair (MMR) (Peltomaki and Vasen, tumours was confirmed by comparisons with 16 tumours 1997). Whilst germline MMR gene mutation is the with low-frequency microsatellite instability and 56 major mechanism responsible for the mismatch repair tumours with stable microsatellites. As anticipated, deficiency in hereditary non-polyposis colorectal cancer patients with family histories usually showed germline (HNPCC) kindreds, the majority of sporadic late-onset mutation of hMSH2 or hMLH1. In many cases the MSI-H colorectal cancer were attributed to hMLH1 residual normal allele was silenced in their tumours by promoter hypermethylation (Kane et al., 1997; Herman loss of heterozygosity (LOH). The small subset of late- et al., 1998; Cunningham et al., 1998; Kuismanen et onset, sporadic cases confirmed the preponderance in this al., 2000). Cancers with MSI-H constitute a high group of biallelic hMLH1 promoter methylation. In the proportion of those arising in the colon and rectum of early-onset, apparently sporadic subset there were 11 younger patients, and whilst many of these have clear tumours with hMLH1 deficiency, five with hMSH2 family histories of cancer, several do not. Relatively deficiency and four with no detectable abnormality in little is known of the molecular origins of the MMR expression of either protein. These showed a complex deficiency in such apparently sporadic early-onset mixture of lesions, including germline and somatic cancers, although we and others have shown that some mutations, promoter methylation, LOH, suppression of arise on a background of germ-line mutation in hMSH-2 or hMLH1, despite the absence of family history (Liu et al., 1995; Chan et al., 1999a). A comprehensive understanding of the mechanisms *Correspondence: SY Leung; E-mail: [email protected] and ST Yuen; E-mail: [email protected] underlying MSI, in particular distinguishing between Received 1 May 2002; revised 8 August 2002; accepted 8 August germline and somatic or epigenetic events, could 2002 significantly influence both the management of the Basis of MSI in colorectal cancer ST Yuen et al 7586 patients and the level of surveillance required for their Figure 1. Methylation-specific PCR, which assessed relatives. There is good reason to believe that the MSI- methylation status of CpG sites located in bases 7694, H phenotype requires bi-allelic (‘two hit’) inactivation 7688, 7674, 7672, 7600, 7598 and 7588 (referred of the responsible MMR gene. In practice, however, to here as region 1), was successfully performed in 40 only a single inactivating event was identified in up to MSI-H, 15 MSI-L and 26 MSS tumours. hMLH1 65% of currently reported MSI-H colorectal tumours promoter methylation was detected in 12 of 22 MSI-H (Hemminki et al., 1994; Konishi et al., 1996; cases with hMLH1 protein loss, but also in several Tannergard et al., 1997; Leung et al., 1998; Chan et tumours in which hMLH1 expression was normal: two al., 1999b). This may reflect the restricted number of of 12 MSI-H cases with hMSH2 protein loss, two of inactivation mechanisms that were directly investi- six MSI-H cases with no MMR protein loss, two of 15 gated. Moreover, there are technical limitations to the MSI-L cases, and zero of 26 MSS cases. The incidence detection of mutations, be they germline or somatic, in of promoter methylation was significantly different the MMR genes. These large genes have no mutation between cases with (12/22) and without (6/59) loss of hot spots and hence no simple strategy can detect all hMLH1 protein (P50.0001), but the clear evidence of mutations. The simple detection of hMLH1 promoter region one methylation in six tumours with normal hypermethylation also suffers from several drawbacks. expression of hMLH1 protein prompted search for Firstly, it does not distinguish between mono-allelic or other promoter regions with which the association of bi-allelic inactivation in primary cancer tissue. methylation and protein expression was more specific. Secondly, the specificity of promoter methylation and When bisulphite sequencing was used to identify hence its functional significance varies according to the methylation in a region more proximal to the methods used and the CpG sites examined (Deng et al., transcription start site, covering bases 7178 to 1999). These limitations are particularly important for 7248 (referred to here as region 2), two distinct the group of sporadic early-onset colorectal cancer patterns emerged. DNA extracted from the tumour with MSI, where so far there is no comprehensive was either unmethylated in all eight CpG sites study addressing the relative contributions of germline, (hereafter referred to as the unmethylated pattern), somatic and epigenetic events. Here we characterize the or showed methylation close to or in excess of 50% of basis of MSI-H in a substantial number of MSI-H total DNA at all eight CpG sites (referred to as the colorectal cancers, including familial, sporadic late- methylated pattern) (Figure 2). Of the six tumours onset and sporadic early-onset cases, the latter group with normal expression of hMLH1 protein but with shows an exceptionally high incidence in Hong Kong methylation in region 1 of the hMLH1 promoter, all (Yuen et al., 1997; Chan et al., 1999a). For both showed an unmethylated pattern in region 2. Of the hMSH2 and hMLH1 we sought to identify loss of 12 tumours with loss of hMLH1 expression and protein expression, germline and somatic mutations, methylated region 1, however, seven also showed the mRNA expression and LOH. Methylation was exam- methylated pattern in region 2. Amongst the remain- ined in various regions of the hMLH1 promoter. The ing five tumours, only two (L6, L20) failed to show data show that different mechanisms of MMR clear evidence of bi-allelic inactivation of hMLH1 by deficiency predominate in these three groups. means other than methylation (i.e. somatic or germ- The results of immunohistochemical staining for line mutation together with LOH), and none failed to hMSH2 and hMLH1, and the family or personal show at least one somatic or germline mutation histories of cancer of 44 individuals with MSI-H (Figure 2B,C). In contrast, of the seven cases with tumours are shown in Table 1 and summarized in region 2 methylation, five showed a predominance of Figure 1. In the majority of these tumours (38/44, methylated DNA in region 2 on bisulphite sequencing 86%) one or other of the hMLH1 or hMSH2 proteins and absence of other germline or somatic inactivation was undetectable. In contrast, both proteins were events. Real time quantitative PCR of these cases readily identified in a nuclear location in all of 16 demonstrated significantly lowered hMLH1 transcript tumours with low levels of microsatellite instability when compared with 10 randomly selected MSS (MSI-L) and 56 tumours with stable microsatellites colorectal cancers (Figure 3). These results strongly (MSS). When the MSI-H tumours were classified on support transcriptional silencing of hMLH1 in these the basis of age and family history into familial cases by bi-allelic promoter methylation in region 2.
Recommended publications
  • Next-Generation Sequencing for Identifying a Novel/De Novo
    Martínez-Hernández et al. BMC Medical Genomics (2019) 12:68 https://doi.org/10.1186/s12920-019-0528-1 CASE REPORT Open Access Next-generation sequencing for identifying a novel/de novo pathogenic variant in a Mexican patient with cystic fibrosis: a case report Angélica Martínez-Hernández1, Julieta Larrosa2, Francisco Barajas-Olmos1, Humberto García-Ortíz1, Elvia C. Mendoza-Caamal3, Cecilia Contreras-Cubas1, Elaheh Mirzaeicheshmeh1, José Luis Lezana4 and Lorena Orozco1* Abstract Background: Mexico is among the countries showing the highest heterogeneity of CFTR variants. However, no de novo variants have previously been reported in Mexican patients with cystic fibrosis (CF). Case presentation: Here, we report the first case of a novel/de novo variant in a Mexican patient with CF. Our patient was an 8-year-old male who had exhibited the clinical onset of CF at one month of age, with steatorrhea, malabsorption, poor weight gain, anemia, and recurrent respiratory tract infections. Complete sequencing of the CFTR gene by next generation sequencing (NGS) revealed two different variants in trans, including the previously reported CF-causing variant c.3266G > A (p.Trp1089*, W1089*), that was inherited from the mother, and the novel/ de novo CFTR variant c.1762G > T (p.Glu588*). Conclusion: Our results demonstrate the efficiency of targeted NGS for making a rapid and precise diagnosis in patients with clinically suspected CF. This method can enable the provision of accurate genetic counselling, and improve our understanding of the molecular basis of genetic diseases. Keywords: Cystic fibrosis, Next generation sequencing, P.Trp1089*, P.Glu588*, Novel/de novo variant Background been detected, with the deletion of phenylalanine at pos- Cystic fibrosis (CF, MIM# 219700) is the most common ition 508 (c.1521_1523delCTT, p.Phe508del, F508del) autosomal recessive disorder among Caucasians.
    [Show full text]
  • Gene Therapy Glossary of Terms
    GENE THERAPY GLOSSARY OF TERMS A • Phase 3: A phase of research to describe clinical trials • Allele: one of two or more alternative forms of a gene that that gather more information about a drug’s safety and arise by mutation and are found at the same place on a effectiveness by studying different populations and chromosome. different dosages and by using the drug in combination • Adeno-Associated Virus: A single stranded DNA virus that has with other drugs. These studies typically involve more not been found to cause disease in humans. This type of virus participants.7 is the most frequently used in gene therapy.1 • Phase 4: A phase of research to describe clinical trials • Adenovirus: A member of a family of viruses that can cause occurring after FDA has approved a drug for marketing. infections in the respiratory tract, eye, and gastrointestinal They include post market requirement and commitment tract. studies that are required of or agreed to by the study • Adeno-Associated Virus Vector: Adeno viruses used as sponsor. These trials gather additional information about a vehicles for genes, whose core genetic material has been drug’s safety, efficacy, or optimal use.8 removed and replaced by the FVIII- or FIX-gene • Codon: a sequence of three nucleotides in DNA or RNA • Amino Acids: building block of a protein that gives instructions to add a specific amino acid to an • Antibody: a protein produced by immune cells called B-cells elongating protein in response to a foreign molecule; acts by binding to the • CRISPR: a family of DNA sequences that can be cleaved by molecule and often making it inactive or targeting it for specific enzymes, and therefore serve as a guide to cut out destruction and insert genes.
    [Show full text]
  • Genomic Profiling Reveals High Frequency of DNA Repair Genetic
    www.nature.com/scientificreports OPEN Genomic profling reveals high frequency of DNA repair genetic aberrations in gallbladder cancer Reham Abdel‑Wahab1,9, Timothy A. Yap2, Russell Madison4, Shubham Pant1,2, Matthew Cooke4, Kai Wang4,5,7, Haitao Zhao8, Tanios Bekaii‑Saab6, Elif Karatas1, Lawrence N. Kwong3, Funda Meric‑Bernstam2, Mitesh Borad6 & Milind Javle1,10* DNA repair gene aberrations (GAs) occur in several cancers, may be prognostic and are actionable. We investigated the frequency of DNA repair GAs in gallbladder cancer (GBC), association with tumor mutational burden (TMB), microsatellite instability (MSI), programmed cell death protein 1 (PD‑1), and its ligand (PD‑L1) expression. Comprehensive genomic profling (CGP) of 760 GBC was performed. We investigated GAs in 19 DNA repair genes including direct DNA repair genes (ATM, ATR , BRCA1, BRCA2, FANCA, FANCD2, MLH1, MSH2, MSH6, PALB2, POLD1, POLE, PRKDC, and RAD50) and caretaker genes (BAP1, CDK12, MLL3, TP53, and BLM) and classifed patients into 3 groups based on TMB level: low (< 5.5 mutations/Mb), intermediate (5.5–19.5 mutations/Mb), and high (≥ 19.5 mutations/Mb). We assessed MSI status and PD‑1 & PD‑L1 expression. 658 (86.6%) had at least 1 actionable GA. Direct DNA repair gene GAs were identifed in 109 patients (14.2%), while 476 (62.6%) had GAs in caretaker genes. Both direct and caretaker DNA repair GAs were signifcantly associated with high TMB (P = 0.0005 and 0.0001, respectively). Tumor PD‑L1 expression was positive in 119 (15.6%), with 17 (2.2%) being moderate or high. DNA repair GAs are relatively frequent in GBC and associated with coexisting actionable mutations and a high TMB.
    [Show full text]
  • Mitosis Vs. Meiosis
    Mitosis vs. Meiosis In order for organisms to continue growing and/or replace cells that are dead or beyond repair, cells must replicate, or make identical copies of themselves. In order to do this and maintain the proper number of chromosomes, the cells of eukaryotes must undergo mitosis to divide up their DNA. The dividing of the DNA ensures that both the “old” cell (parent cell) and the “new” cells (daughter cells) have the same genetic makeup and both will be diploid, or containing the same number of chromosomes as the parent cell. For reproduction of an organism to occur, the original parent cell will undergo Meiosis to create 4 new daughter cells with a slightly different genetic makeup in order to ensure genetic diversity when fertilization occurs. The four daughter cells will be haploid, or containing half the number of chromosomes as the parent cell. The difference between the two processes is that mitosis occurs in non-reproductive cells, or somatic cells, and meiosis occurs in the cells that participate in sexual reproduction, or germ cells. The Somatic Cell Cycle (Mitosis) The somatic cell cycle consists of 3 phases: interphase, m phase, and cytokinesis. 1. Interphase: Interphase is considered the non-dividing phase of the cell cycle. It is not a part of the actual process of mitosis, but it readies the cell for mitosis. It is made up of 3 sub-phases: • G1 Phase: In G1, the cell is growing. In most organisms, the majority of the cell’s life span is spent in G1. • S Phase: In each human somatic cell, there are 23 pairs of chromosomes; one chromosome comes from the mother and one comes from the father.
    [Show full text]
  • Human Germline Genome Editing: Fact Sheet
    Human germline genome editing: fact sheet Purpose • To contribute to evidence-informed discussions about human germline genome editing. KEY TAKEAWAYS • Gene editing offers the potential to improve human health in ways not previously possible. • Making changes to human genes that can be passed on to future generations is prohibited in Australia. • Unresolved questions remain on the possible long-term impacts, unintended consequences, and ethical issues associated with introducing heritable changes by editing of the genome of human gametes (sperm and eggs) and embryos. • AusBiotech believes the focus of human gene editing should remain on non-inheritable changes until such time as the scientific evidence, regulatory frameworks and health care models have progressed sufficiently to warrant consideration of any heritable genetic edits. Gene editing Gene editing is the insertion, deletion, or modification of DNA to modify an organism’s specific genetic characteristics. New and evolving gene editing techniques and tools (e.g. CRISPR) allow editing of genes with a level of precision that increases its applications across the health, agricultural, and industrial sectors. These breakthrough techniques potentially offer a range of different options for treating devastating human diseases and delivering environmentally sustainable food production systems that can feed the world’s growing population, which is expected to exceed nine billion by 2050. The current primary application of human gene editing is on non-reproductive cells (‘somatic’ cells)
    [Show full text]
  • Exploring the Interplay of Telomerase Reverse Transcriptase and Β-Catenin in Hepatocellular Carcinoma
    cancers Review Exploring the Interplay of Telomerase Reverse Transcriptase and β-Catenin in Hepatocellular Carcinoma Srishti Kotiyal and Kimberley Jane Evason * Department of Oncological Sciences, Department of Pathology, and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-801-587-4606 Simple Summary: Liver cancer is one of the deadliest human cancers. Two of the most common molecular aberrations in liver cancer are: (1) activating mutations in the gene encoding β-catenin (CTNNB1); and (2) promoter mutations in telomerase reverse transcriptase (TERT). Here, we review recent findings regarding the interplay between TERT and β-catenin in order to better understand their role in liver cancer. Abstract: Hepatocellular carcinoma (HCC) is one of the deadliest human cancers. Activating muta- tions in the telomerase reverse transcriptase (TERT) promoter (TERTp) and CTNNB1 gene encoding β-catenin are widespread in HCC (~50% and ~30%, respectively). TERTp mutations are predicted to increase TERT transcription and telomerase activity. This review focuses on exploring the role of TERT and β-catenin in HCC and the current findings regarding their interplay. TERT can have contradictory effects on tumorigenesis via both its canonical and non-canonical functions. As a critical regulator of proliferation and differentiation in progenitor and stem cells, activated β-catenin drives HCC; however, inhibiting endogenous β-catenin can also have pro-tumor effects. Clinical studies revealed a significant concordance between TERTp and CTNNB1 mutations in HCC. In Citation: Kotiyal, S.; Evason, K.J. stem cells, TERT acts as a co-factor in β-catenin transcriptional complexes driving the expression Exploring the Interplay of Telomerase of WNT/β-catenin target genes, and β-catenin can bind to the TERTp to drive its transcription.
    [Show full text]
  • Telomere and Telomerase in Oncology
    Cell Research (2002); 12(1):1-7 http://www.cell-research.com REVIEW Telomere and telomerase in oncology JIAO MU*, LI XIN WEI International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China ABSTRACT Shortening of the telomeric DNA at the chromosome ends is presumed to limit the lifespan of human cells and elicit a signal for the onset of cellular senescence. To continually proliferate across the senescent checkpoint, cells must restore and preserve telomere length. This can be achieved by telomerase, which has the reverse transcriptase activity. Telomerase activity is negative in human normal somatic cells but can be detected in most tumor cells. The enzyme is proposed to be an essential factor in cell immortalization and cancer progression. In this review we discuss the structure and function of telomere and telomerase and their roles in cell immortalization and oncogenesis. Simultaneously the experimental studies of telomerase assays for cancer detection and diagnosis are reviewed. Finally, we discuss the potential use of inhibitors of telomerase in anti-cancer therapy. Key words: Telomere, telomerase, cancer, telomerase assay, inhibitor. Telomere and cell replicative senescence base pairs of the end of telomeric DNA with each Telomeres, which are located at the end of round of DNA replication. Hence, the continual chromosome, are crucial to protect chromosome cycles of cell growth and division bring on progress- against degeneration, rearrangment and end to end ing telomere shortening[4]. Now it is clear that te- fusion[1]. Human telomeres are tandemly repeated lomere shortening is responsible for inducing the units of the hexanucleotide TTAGGG. The estimated senescent phenotype that results from repeated cell length of telomeric DNA varies from 2 to 20 kilo division, but the mechanism how a short telomere base pairs, depending on factors such as tissue type induces the senescence is still unknown.
    [Show full text]
  • Multi-Omics Analysis Defines Core Genomic Alterations in Pheochromocytomas and Paragangliomas
    ARTICLE Received 11 Aug 2014 | Accepted 5 Dec 2014 | Published 27 Jan 2015 DOI: 10.1038/ncomms7044 OPEN Multi-omics analysis defines core genomic alterations in pheochromocytomas and paragangliomas Luis Jaime Castro-Vega1,2,*, Eric Letouze´3,*, Nelly Burnichon1,2,4,*, Alexandre Buffet1,2,4, Pierre-He´lie Disderot1,2, Emmanuel Khalifa1,2,4,Ce´line Loriot1,2, Nabila Elarouci3, Aure´lie Morin1,2,Me´lanie Menara1,2, Charlotte Lepoutre-Lussey1,2,5,Ce´cile Badoual1,2,6, Mathilde Sibony2,7, Bertrand Dousset2,8,9,10, Rossella Libe´2,9,10,11,12, Franck Zinzindohoue2,13, Pierre Franc¸ois Plouin1,2,5,12,Je´roˆme Bertherat2,9,10,11,12, Laurence Amar1,2,5, Aure´lien de Reynie`s3, Judith Favier1,2,y & Anne-Paule Gimenez-Roqueplo1,2,4,12,y Pheochromocytomas and paragangliomas (PCCs/PGLs) are neural crest-derived tumours with a very strong genetic component. Here we report the first integrated genomic examination of a large collection of PCC/PGL. SNP array analysis reveals distinct copy-number patterns associated with genetic background. Whole-exome sequencing shows a low mutation rate of 0.3 mutations per megabase, with few recurrent somatic mutations in genes not previously associated with PCC/PGL. DNA methylation arrays and miRNA sequencing identify DNA methylation changes and miRNA expression clusters strongly associated with messenger RNA expression profiling. Overexpression of the miRNA cluster 182/96/183 is specific in SDHB-mutated tumours and induces malignant traits, whereas silencing of the imprinted DLK1-MEG3 miRNA cluster appears as a potential driver in a subgroup of sporadic tumours.
    [Show full text]
  • High-Efficiency CRISPR/Cas9 Mutagenesis of the White Gene in the Milkweed Bug Oncopeltus Fasciatus
    | INVESTIGATION High-Efficiency CRISPR/Cas9 Mutagenesis of the white Gene in the Milkweed Bug Oncopeltus fasciatus Katie Reding and Leslie Pick1 Department of Entomology, University of Maryland, College Park, Maryland 20742 ORCID IDs: 0000-0003-2067-4232 (K.R.); 0000-0002-4505-5107 (L.P.) ABSTRACT In this manuscript, we report that clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 is highly efficient in the hemipteran Oncopeltus fasciatus. The white gene is well characterized in Drosophila where mutation causes loss of eye pigmentation; white is a reliable marker for transgenesis and other genetic manipulations. Accordingly, white has been targeted in a number of nonmodel insects to establish tools for genetic studies. Here, we generated mutations in the Of-white (Of-w) locus using CRISPR/Cas9. We found that Of-w is required for pigmentation throughout the body of Oncopeltus, not just the ommatidia. High rates of somatic mosaicism were observed in the injected generation, reflecting biallelic mutations, and a high rate of germline mutation was evidenced by the large proportion of heterozygous G1s. However, Of-w mutations are homozygous lethal; G2 homozygotes lacked pigment dispersion throughout the body and did not hatch, precluding the establishment of a stable mutant line. Embryonic and parental RNA interference (RNAi) were subsequently performed to rule out off-target mutations producing the observed phenotype and to evaluate the efficacy of RNAi in ablating gene function compared to a loss-of-function mutation. RNAi knockdowns phe- nocopied Of-w homozygotes, with an unusual accumulation of orange granules observed in unhatched embryos. This is, to our knowledge, the first CRISPR/Cas9-targeted mutation generated in Oncopeltus.
    [Show full text]
  • Cfdna Deconvolution Via NIPT of a Pregnant Woman After Bone Marrow
    Zhu et al. Human Genomics (2021) 15:14 https://doi.org/10.1186/s40246-021-00311-w PRIMARY RESEARCH Open Access cfDNA deconvolution via NIPT of a pregnant woman after bone marrow transplant and donor egg IVF Jianjiang Zhu1 , Feng Hui2, Xuequn Mao1, Shaoqin Zhang1, Hong Qi1* and Yang Du2* Abstract Cell-free DNA is known to be a mixture of DNA fragments originating from various tissue types and organs of the human body and can be utilized for several clinical applications and potentially more to be created. Non-invasive prenatal testing (NIPT), by high throughput sequencing of cell-free DNA (cfDNA), has been successfully applied in the clinical screening of fetal chromosomal aneuploidies, with more extended coverage under active research. In this study, via a quite unique and rare NIPT sample, who has undergone both bone marrow transplant and donor egg IVF, we investigated the sources of oddness observed in the NIPT result using a combination of molecular genetics and genomic methods and eventually had the case fully resolved. Along the process, we devised a clinically viable process to dissect the sample mixture. Eventually, we used the proposed scheme to evaluate the relatedness of individuals and the demultiplexed sample components following modified population genetics concepts, exemplifying a noninvasive prenatal paternity test prototype. For NIPT specific applicational concern, more thorough and detailed clinical information should therefore be collected prior to cfDNA-based screening procedure like NIPT and systematically reviewed when an abnormal report is obtained to improve genetic counseling and overall patient care. Keywords: NIPT, Target sequencing, Fetal fraction, IVF, Transplant, Prenatal diagnostic Introduction establishment of circulating tumor DNA (ctDNA) in the Cell-free DNA (cfDNA) is known to be a mixture from plasma of cancer patients [4].
    [Show full text]
  • Genetic Testing for Reproductive Carrier Screening and Prenatal Diagnosis
    Medical Coverage Policy Effective Date ............................................. 7/15/2021 Next Review Date ......................................12/15/2021 Coverage Policy Number .................................. 0514 Genetic Testing for Reproductive Carrier Screening and Prenatal Diagnosis Table of Contents Related Coverage Resources Overview ........................................................ 2 Genetics Coverage Policy ............................................ 2 Genetic Testing Collateral File Genetic Counseling ...................................... 2 Recurrent Pregnancy Loss: Diagnosis and Treatment Germline Carrier Testing for Familial Infertility Services Disease .......................................................... 3 Preimplantation Genetic Testing of an Embryo........................................................... 4 Preimplantation Genetic Testing (PGT-A) .. 5 Sequencing–Based Non-Invasive Prenatal Testing (NIPT) ............................................... 5 Invasive Prenatal Testing of a Fetus .......... 6 Germline Mutation Reproductive Genetic Testing for Recurrent Pregnancy Loss ...... 6 Germline Mutation Reproductive Genetic Testing for Infertility ..................................... 7 General Background .................................... 8 Genetic Counseling ...................................... 8 Germline Genetic Testing ............................ 8 Carrier Testing for Familial Disease ........... 8 Preimplantation Genetic Testing of an Embryo..........................................................
    [Show full text]
  • Intercellular Competition and the Inevitability of Multicellular Aging
    Intercellular competition and the inevitability of multicellular aging Paul Nelsona,1 and Joanna Masela aDepartment of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ 85721 Edited by Raghavendra Gadagkar, Indian Institute of Science, Bangalore, India, and approved October 6, 2017 (received for review November 14, 2016) Current theories attribute aging to a failure of selection, due to Aging in multicellular organisms occurs at both the cellular either pleiotropic constraints or declining strength of selection and intercellular levels (17). Multicellular organisms, by definition, after the onset of reproduction. These theories implicitly leave require a high degree of intercellular cooperation to maintain open the possibility that if senescence-causing alleles could be homeostasis. Often, cellular traits required for producing a viable identified, or if antagonistic pleiotropy could be broken, the multicellular phenotype come at a steep cost to individual cells effects of aging might be ameliorated or delayed indefinitely. (14, 18, 19). Conversely, many mutant cells that do not invest in These theories are built on models of selection between multicel- holistic organismal fitness have a selective advantage over cells lular organisms, but a full understanding of aging also requires that do. If intercellular competition occurs, such “cheater” or examining the role of somatic selection within an organism. “defector” cells may proliferate and displace “cooperating” cells, Selection between somatic cells (i.e., intercellular competition) with detrimental consequences for the multicellular organism can delay aging by purging nonfunctioning cells. However, the (20, 21). Cancer, a leading cause of death in humans at rates that fitness of a multicellular organism depends not just on how increase with age, is one obvious manifestation of cheater pro- functional its individual cells are but also on how well cells work liferation (22–24).
    [Show full text]