Structural and Functional Consequences of a Disease Mutation in the Telomere Protein TPP1

Total Page:16

File Type:pdf, Size:1020Kb

Structural and Functional Consequences of a Disease Mutation in the Telomere Protein TPP1 Structural and functional consequences of a disease mutation in the telomere protein TPP1 Kamlesh Bishta,1, Eric M. Smitha,b,1, Valerie M. Tesmera, and Jayakrishnan Nandakumara,b,2 aDepartment of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109; and bProgram in Chemical Biology, University of Michigan, Ann Arbor, MI 48109 Edited by Joachim Lingner, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland, and accepted by Editorial Board Member Dinshaw J. Patel September 29, 2016 (received for review April 8, 2016) Telomerase replicates chromosome ends to facilitate continued cell The discovery of the TEL patch of TPP1 prompted the pre- division. Mutations that compromise telomerase function result in diction that this region could be a hotspot for mutations that stem cell failure diseases, such as dyskeratosis congenita (DC). One cause telomerase-deficiency diseases, such as DC. We recently such mutation (K170Δ), residing in the telomerase-recruitment factor reported a case of a severe variant of DC, Hoyeraal–Hreidarsson TPP1, provides an excellent opportunity to structurally, biochemi- syndrome (23), in which the proband was heterozygous for a cally, and genetically dissect the mechanism of such diseases. We deletion of a single amino acid of the TPP1 protein, namely lysine ACD show through site-directed mutagenesis and X-ray crystallography 170 (K170) (24). Our study placed the gene coding for TPP1 DKC1 TERC TERT that this TPP1 disease mutation deforms the conformation of two protein on a list with 10 other genes ( , , , RTEL1 TINF2 CTC1 NOP10 NHP2 WRAP53 PARN critical amino acids of the TEL [TPP1’s glutamate (E) and leucine-rich , , , , , ,and ) that are found mutated in DC and other telomere-related dis- (L)] patch, the surface of TPP1 that binds telomerase. Using CRISPR- Δ Cas9 technology, we demonstrate that introduction of this mutation orders (25). Indeed, a heterozygous TPP1 K170 mutation was in a heterozygous manner is sufficient to shorten telomeres in human also reported in another unrelated family, where it was implicated in causing aplastic anemia and other related hematopoietic com- cells. Our findings rule out dominant-negative effects of the muta- plications in the proband (26). In both families, the presence of the tion. Instead, these findings implicate reduced TEL patch dosage in K170Δ mutation correlated strongly with short telomeres. Tran- causing telomere shortening. Our studies provide mechanistic insight sient overexpression of K170Δ in cultured human cells resulted in a into telomerase-deficiency diseases and encourage the development decrease in telomerase recruitment (24, 26), and a reduction in the of gene therapies to counter such diseases. ability of TPP1 to stimulate telomerase processivity (the ability of telomerase to continue DNA synthesis without dissociating from a telomere | telomerase | TPP1 | TEL patch | dyskeratosis congenita bound primer) (24). These results invoke a direct role for the K170Δ mutation in reducing telomerase function in dividing cells. elomerase is a unique ribonuclear protein complex [telomerase However, it is unknown whether the heterozygous K170Δ mutation Treverse transcriptase (TERT): protein subunit; TR: RNA sub- is sufficient to cause telomere shortening in dividing human cells unit] that synthesizes telomeric DNA repeats (GGTTAG in hu- without additional genetic modifiers. BIOCHEMISTRY mans) at chromosome ends to facilitate continued cell division The placement of K170 directly adjacent to E168, E169, and (1–3). Although telomerase is not expressed in most normal so- E171 strongly suggests that this amino acid facilitates the matic cells, its presence in stem cells is crucial for their self-renewal ability of the TEL patch to recognize telomerase. However, (4). Germ-line mutations in the genes coding for telomerase sub- inspection of the crystal structure of the OB domain of WT units, or in protein factors important for either telomerase bio- genesis or trafficking in the cell, result in genetically inherited Significance diseases, the most prominent of which is dyskeratosis congenita (DC) (5, 6). DC is clinically diagnosed by the presentation of a Telomerase is an enzyme that replicates chromosome ends to classic triad: nail dysplasia, skin hyperpigmentation, and oral leu- facilitate continued stem cell division. Mutations in telomerase or koplakia. Bone marrow failure resulting from the depletion of the in telomerase-related genes result in stem cell-dysfunction dis- hematopoietic stem cell pool is the primary cause of morbidity and eases, such as dyskeratosis congenita (DC). Despite its devastat- mortality in DC patients. At a molecular level, DC is diagnosed by ing nature, DC currently has no cure. Here we report the crystal blood leukocyte telomere lengths of less than the first percentile structure of a mutant protein implicated in DC to reveal how the for age. Despite the devastating nature of this disease, its genetic mutation disrupts a region of the protein essential for telomer- basis has hampered the development of effective therapies. ase function. Furthermore, we demonstrated that this mutation, The TPP1 (TINT1/PTOP/PIP1) protein (7–9) is a component when introduced into a human cell line, is sufficient to cause the of the six-membered shelterin complex [TPP1, protection of telo- cellular underpinnings of DC. Our results therefore make the meres protein (POT1), telomeric repeat-binding factor 1 (TRF1), strong prediction that correcting the mutation in the stem cells of TRF2, Rap1, and TERF1-interacting nuclear factor 2 (TIN2)] the patient will reverse the cellular symptoms of disease. that protects chromosome ends from the cellular DNA damage response and repair machineries (10, 11). TPP1 is a unique Author contributions: K.B., E.M.S., and J.N. designed research; K.B., E.M.S., V.M.T., and shelterin component in that it binds telomerase (12), recruits J.N. performed research; K.B., E.M.S., V.M.T., and J.N. analyzed data; and J.N. wrote telomerase to telomeres (13), increases telomerase processivity the paper. (14), and facilitates telomere elongation in telomerase-positive The authors declare no conflict of interest. cells (15, 16). Mutagenesis screens revealed a region called the This article is a PNAS Direct Submission. J.L. is a Guest Editor invited by the Editorial TEL [TPP1’s glutamate (E) and leucine-rich (L)] patch, which Board. resides in the structurally characterized oligosaccharide– Freely available online through the PNAS open access option. oligonucleotide-binding (OB) domain of the TPP1 protein. The Data deposition: The atomic coordinates and structure factors have been deposited in the TEL patch is critical for all of the telomerase-associated roles of Protein Data Bank, www.pdb.org (PDB ID codes 5I2X and 5I2Y). TPP1 (15–22). Central to TEL patch function are three glutamates— 1K.B. and E.M.S. contributed equally to this work. E168, E169, and E171 (Fig. 1A)—which, when mutated to ala- 2To whom correspondence should be addressed. Email: [email protected]. nine (15, 18) or arginine (17), result in severely compromised This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. telomerase function. 1073/pnas.1605685113/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1605685113 PNAS | November 15, 2016 | vol. 113 | no. 46 | 13021–13026 Downloaded by guest on September 23, 2021 A C D Results Deletion of K170, but Not Its Substitution to Alanine, Greatly Reduces Telomerase Processivity. The deleterious effect of the TPP1 K170Δ mutation on telomerase processivity is not because of a defect in binding to POT1 as K170Δ binds POT1 similar to WT TPP1 protein (Fig. S1A). To further understand the molecular basis of telomerase deficiency caused by this DC mutation, we engineered B a K170A mutation in the TPP1-N vector (14) for expression in Escherichia coli. TPP1-N constructs (TPP1 amino acids 90–334) of K170A, WT, K170Δ, and E169A/E171A (EE-AA) proteins E were purified as described previously (14, 15) (Fig. S1B). We performed direct primer extension assays to determine how each of the TPP1-N variants contributes to telomerase processivity in the presence of POT1 (Fig. 1 B and C). The WT protein displayed the expected increase in telomerase processivity (14), giving rise to a large fraction of longer DNA products. Both K170Δ and E169A/ E171A failed to increase telomerase processivity to WT TPP1-N levels, as expected from previous studies (Fig. 1 B,andC) (15, 24). Strikingly, mutation of K170 to alanine resulted in only a modest reduction in telomerase processivity (Fig. 1 B and C) that was not statistically significant in experiments performed in triplicate (P = F 0.08). This result suggests that the positive charge of K170 is not critical for telomerase processivity. In addition, E169D/E171D (E→D mutation that retain negative charge), but not E169Q/ E171Q (E→Q mutations that are iso-steric and polar but lack the negative charge), was able to stimulate telomerase processivity (Fig. S1C). These results are fully consistent with the structure of TPP1–OB WT, which indicates that the glutamates, but not K170, of the TEL patch loop are accessible to telomerase. Fig. 1. The main-chain, but not the side-chain, of TPP1 K170 is important for Deletion of K170, but Not Its Substitution to Alanine, Greatly Reduces stimulating telomerase processivity, recruiting telomerase to telomeres, and Telomerase Recruitment to Telomeres and Telomere Lengthening. for telomere elongation. (A) Ribbon depiction of a part of the TPP1–OB WT Given that deletion of K170 reduces telomerase processivity but crystal structure (PDB ID code 2I46) with the TEL patch loop (amino acids substitution of K170 to alanine does not, we next asked how K170A 166–171) shown in stick representation. The protrusion in the loop is referred impacts telomerase recruitment. To this end, we engineered HeLa to here as a “knuckle.” (B) Direct primer extension assays with telomerase cell lines stably expressing either FLAG-TPP1 K170A or FLAG- extracts performed in the presence of purified POT1 (500 nM) and the in- TPP1 K170Δ protein using the Flp-recombinase–based single-site dicated TPP1-N proteins (500 nM).
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]
  • Pulmonary Fibrosis Associated with TINF2 Gene Mutation: Is Somatic Reversion Required?
    Pulmonary fibrosis associated with TINF2 gene mutation: is somatic reversion required? To the Editor: We read with great interest the case reported by FUKUHARA et al. [1] of a 43-year-old female patient with dyskeratosis congenita, pulmonary fibrosis and heterozygous mutation in TINF2 (telomerase repeat binding factor 1-interacting nuclear factor 2). TIN2, the TINF2 gene product, TERT (telomere reverse transcriptase) and TERC (telomerase RNA component) participate in the regulation of telomere elongation, in which mutations have been previously found to be associated with familial pulmonary fibrosis in adults [2]. Indeed mutations of SFTPC, coding for surfactant protein C, were initially described in children before being described in adults as old as 72 years of age who presented with familial pulmonary fibrosis [3]. However, we were surprised that a TINF2 mutation could be evidenced in an adult of that age. As highlighted by FUKUHARA et al. [1], patients with the TINF2 mutation present with severe haematological symptoms before 10 years of age [4]. As mentioned by FUKUHARA et al. [1], the identified mutation is probably not hypomorphic because it is a frame-shift deletion located in the mutational ‘‘hot spot’’ described previously. Furthermore, the patient presented with very short telomeres. The TINF2 mutation was probably inherited from her father because he had abnormal skin pigmentation and aplastic anaemia [1]. Re-analysis of the gene mutation sequencing could provide new hypotheses for this late disease onset. Indeed, the electrophoregram depicted in figure 1b in the study by FUKUHARA et al. [1] probably comes from a PCR product sub-cloned into an expression vector [5], and does not ensure that the deletion is at the heterozygous status usually seen in our patients (fig.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Genetics of Familial Non-Medullary Thyroid Carcinoma (FNMTC)
    cancers Review Genetics of Familial Non-Medullary Thyroid Carcinoma (FNMTC) Chiara Diquigiovanni * and Elena Bonora Unit of Medical Genetics, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; [email protected] * Correspondence: [email protected]; Tel.: +39-051-208-8418 Simple Summary: Non-medullary thyroid carcinoma (NMTC) originates from thyroid follicular epithelial cells and is considered familial when occurs in two or more first-degree relatives of the patient, in the absence of predisposing environmental factors. Familial NMTC (FNMTC) cases show a high genetic heterogeneity, thus impairing the identification of pivotal molecular changes. In the past years, linkage-based approaches identified several susceptibility loci and variants associated with NMTC risk, however only few genes have been identified. The advent of next-generation sequencing technologies has improved the discovery of new predisposing genes. In this review we report the most significant genes where variants predispose to FNMTC, with the perspective that the integration of these new molecular findings in the clinical data of patients might allow an early detection and tailored therapy of the disease, optimizing patient management. Abstract: Non-medullary thyroid carcinoma (NMTC) is the most frequent endocrine tumor and originates from the follicular epithelial cells of the thyroid. Familial NMTC (FNMTC) has been defined in pedigrees where two or more first-degree relatives of the patient present the disease in absence of other predisposing environmental factors. Compared to sporadic cases, FNMTCs are often multifocal, recurring more frequently and showing an early age at onset with a worse outcome. FNMTC cases Citation: Diquigiovanni, C.; Bonora, E.
    [Show full text]
  • Produktinformation
    Produktinformation Diagnostik & molekulare Diagnostik Laborgeräte & Service Zellkultur & Verbrauchsmaterial Forschungsprodukte & Biochemikalien Weitere Information auf den folgenden Seiten! See the following pages for more information! Lieferung & Zahlungsart Lieferung: frei Haus Bestellung auf Rechnung SZABO-SCANDIC Lieferung: € 10,- HandelsgmbH & Co KG Erstbestellung Vorauskassa Quellenstraße 110, A-1100 Wien T. +43(0)1 489 3961-0 Zuschläge F. +43(0)1 489 3961-7 [email protected] • Mindermengenzuschlag www.szabo-scandic.com • Trockeneiszuschlag • Gefahrgutzuschlag linkedin.com/company/szaboscandic • Expressversand facebook.com/szaboscandic TNKS2 Antibody, HRP conjugated Product Code CSB-PA867136LB01HU Abbreviation Tankyrase-2 Storage Upon receipt, store at -20°C or -80°C. Avoid repeated freeze. Uniprot No. Q9H2K2 Immunogen Recombinant Human Tankyrase-2 protein (1-246AA) Raised In Rabbit Species Reactivity Human Tested Applications ELISA Relevance Poly-ADP-ribosyltransferase involved in various processes such as Wnt signaling pathway, telomere length and vesicle trafficking. Acts as an activator of the Wnt signaling pathway by mediating poly-ADP-ribosylation of AXIN1 and AXIN2, 2 key components of the beta-catenin destruction complex: poly-ADP- ribosylated target proteins are recognized by RNF146, which mediates their ubiquitination and subsequent degradation. Also mediates poly-ADP-ribosylation of BLZF1 and CASC3, followed by recruitment of RNF146 and subsequent ubiquitination. Mediates poly-ADP-ribosylation of TERF1, thereby contributing
    [Show full text]
  • Rat Anti-TERF1 Monoclonal Antibody, Clone 683D (CABT-RM172) This Product Is for Research Use Only and Is Not Intended for Diagnostic Use
    Rat Anti-TERF1 monoclonal antibody, clone 683D (CABT-RM172) This product is for research use only and is not intended for diagnostic use. PRODUCT INFORMATION Specificity Specifically detects murine Telomeric repeat-binding factor 1 (TRF1). Target TERF1 Immunogen His-tagged full-length recombinant mouse Telomeric repeat-binding factor 1 (TRF1). Isotype IgG1, κ Source/Host Rat Species Reactivity Mouse Clone 683D Purification Protein G purified Conjugate unconjugated Applications FC, ICC, IF, WB Molecular Weight ~51 kDa observed; 48.22 kDa calculated. Uncharacterized bands may be observed in some lysate(s). Format Liquid Size 100 μg, 25 μg Buffer 0.1 M Tris-Glycine (pH 7.4), 150 mM NaCl Preservative 0.05% sodium azide Storage Stable for 1 year at 2-8°C from date of receipt. Warnings Unless otherwise stated in our catalog or other company documentation accompanying the product(s), our products are intended for research use only and are not to be used for any other purpose, which includes but is not limited to, unauthorized commercial uses, in vitro diagnostic uses, ex vivo or in vivo therapeutic uses or any type of consumption or application to humans or animals. 45-1 Ramsey Road, Shirley, NY 11967, USA Email: [email protected] Tel: 1-631-624-4882 Fax: 1-631-938-8221 1 © Creative Diagnostics All Rights Reserved BACKGROUND Introduction Telomeric repeat-binding factor 1 is encoded by the Terf1 gene in murine species. TRF1 is a component of the shelterin complex that is involved in the regulation of telomere length and protection. It binds to telomeric DNA as a homodimer and protects telomeres.
    [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]
  • The Genetics and Clinical Manifestations of Telomere Biology Disorders Sharon A
    REVIEW The genetics and clinical manifestations of telomere biology disorders Sharon A. Savage, MD1, and Alison A. Bertuch, MD, PhD2 3 Abstract: Telomere biology disorders are a complex set of illnesses meric sequence is lost with each round of DNA replication. defined by the presence of very short telomeres. Individuals with classic Consequently, telomeres shorten with aging. In peripheral dyskeratosis congenita have the most severe phenotype, characterized blood leukocytes, the cells most extensively studied, the rate 4 by the triad of nail dystrophy, abnormal skin pigmentation, and oral of attrition is greatest during the first year of life. Thereafter, leukoplakia. More significantly, these individuals are at very high risk telomeres shorten more gradually. When the extent of telo- of bone marrow failure, cancer, and pulmonary fibrosis. A mutation in meric DNA loss exceeds a critical threshold, a robust anti- one of six different telomere biology genes can be identified in 50–60% proliferative signal is triggered, leading to cellular senes- of these individuals. DKC1, TERC, TERT, NOP10, and NHP2 encode cence or apoptosis. Thus, telomere attrition is thought to 1 components of telomerase or a telomerase-associated factor and TINF2, contribute to aging phenotypes. 5 a telomeric protein. Progressively shorter telomeres are inherited from With the 1985 discovery of telomerase, the enzyme that ex- generation to generation in autosomal dominant dyskeratosis congenita, tends telomeric nucleotide repeats, there has been rapid progress resulting in disease anticipation. Up to 10% of individuals with apparently both in our understanding of basic telomere biology and the con- acquired aplastic anemia or idiopathic pulmonary fibrosis also have short nection of telomere biology to human disease.
    [Show full text]
  • Structural Genomics Approach to Investigate Deleterious Impact Of
    www.nature.com/scientificreports OPEN Structural genomics approach to investigate deleterious impact of nsSNPs in conserved telomere maintenance component 1 Arunabh Choudhury1,5, Taj Mohammad2,5, Nikhil Samarth3, Afzal Hussain4, Md. Tabish Rehman4, Asimul Islam2, Mohamed F. Alajmi4, Shailza Singh3 & Md. Imtaiyaz Hassan2* Conserved telomere maintenance component 1 (CTC1) is an important component of the CST (CTC1-STN1-TEN1) complex, involved in maintaining the stability of telomeric DNA. Several non- synonymous single-nucleotide polymorphisms (nsSNPs) in CTC1 have been reported to cause Coats plus syndrome and Dyskeratosis congenital diseases. Here, we have performed sequence and structure analyses of nsSNPs of CTC1 using state-of-the-art computational methods. The structure- based study focuses on the C-terminal OB-fold region of CTC1. There are 11 pathogenic mutations identifed, and detailed structural analyses were performed. These mutations cause a signifcant disruption of noncovalent interactions, which may be a possible reason for CTC1 instability and consequent diseases. To see the impact of such mutations on the protein conformation, all-atom molecular dynamics (MD) simulations of CTC1-wild-type (WT) and two of the selected mutations, R806C and R806L for 200 ns, were carried out. A signifcant conformational change in the structure of the R806C mutant was observed. This study provides a valuable direction to understand the molecular basis of CTC1 dysfunction in disease progression, including Coats plus syndrome. Unlike prokaryotic chromosomes, eukaryotic chromosomes are linear and are much larger in size. Te ends of the eukaryotic chromosome are composed of a specialized protein-DNA complex called telomeres which maintains the stability of the chromosome ends1.
    [Show full text]