GENETICS IN ENDOCRINOLOGY Genetic Diagnosis Of . - Bioscientifica

1y ago
11 Views
3 Downloads
1,008.44 KB
13 Pages
Last View : 1m ago
Last Download : 3m ago
Upload by : Elisha Lemon
Transcription

ReviewL Persani and othersNGS in endocrine diseases179:3R111–R123GENETICS IN ENDOCRINOLOGYGenetic diagnosis of endocrine diseases byNGS: novel scenarios and unpredictableresults and risksLuca Persani1,2, Tiziana de Filippis2, Carla Colombo2 and Davide Gentilini3,41Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy, Labs of 2Endocrine andMetabolic Research, 3Molecular Biology Research, IRCCS Istituto Auxologico Italiano, Milan, Italy, and 4University ofPavia, Pavia, ItalyCorrespondenceshould be addressedto L PersaniEmailluca.persani@unimi.itEuropean Journal of EndocrinologyAbstractThe technological advancements in genetics produced a profound impact on the research and diagnostics of noncommunicable diseases. The availability of next-generation sequencing (NGS) allowed the identification of novelcandidate genes but also an in-depth modification of the understanding of the architecture of several endocrinediseases. Several different NGS approaches are available allowing the sequencing of several regions of interest orthe whole exome or genome (WGS, WES or targeted NGS), with highly variable costs, potentials and limitationsthat should be clearly known before designing the experiment. Here, we illustrate the NGS scenario, describe theadvantages and limitations of the different protocols and review some of the NGS results obtained in differentendocrine conditions. We finally give insights on the terminology and requirements for the implementation of NGS inresearch and diagnostic labs.European Journal ofEndocrinology(2018) 179, R111–R123IntroductionThe technological advancements in genetics have hada profound impact on the research and diagnostics ofnon-communicable diseases. In several of these cases,the availability of next-generation sequencing (NGS)allowed the identification of novel candidate genes butalso an in-depth modification of the understanding of thearchitecture of several diseases. Thanks to the power ofthese approaches and the progressive diminution of costs,Invited author’s profileDr L Persani is Full Professor of Endocrinology at the Department of Clinical Sciences& Community Health of the University of Milan and is the Chief of the Department ofEndocrinologyand Metabolic Diseases at the Istituto Auxologico Italiano (a nonprofit Institutionfor Research and Cure supported by the Italian Ministry of Health) in Milan, Italy. Dr L Persaniis a translational researcher and has been leading a Research Lab that provides molecular testingand familial counselling for rare heritable endocrine diseases. In recent years, he developed NGStargeted analyses for disorders of the thyroid, pituitary, adrenals, growth, sex development andmaturation, as well as for multiple endocrine 0/EJE-18-0379 2018 European Society of EndocrinologyPrinted in Great BritainPublished by Bioscientifica Ltd.Downloaded from Bioscientifica.com at 05/17/2022 12:43:34AMvia free access

ReviewL Persani and othersthese changes have been occurring in a short period of timeand a large portion of clinicians are not prepared for suchrevolution, and misinterpretation of the NGS informationrepresents a real danger. This manuscript illustrates thenew-generation approaches for DNA sequencing and thenovel scenarios that we are currently facing in order tomake the clinical endocrinologists aware of the potentialadvantages and risks of this revolution in genetics.European Journal of EndocrinologyVariable approaches: NGS panel/targetedanalyses vs WES or WGSNGS is being adopted by genome diagnostics laboratoriesin different countries worldwide. However, implementingNGS-based tests according to diagnostic standards isa challenge for individual laboratories (1). One of themost important issues that need to be addressed regardsthe selection of the library to be adopted. The mainquestion is: ‘Should I make a custom panel for this geneset, or should I do whole exome sequencing (WES), or isit better to perform whole genome sequencing (WGS)?’The question is crucial since several issues of diagnosticor research workflow will depend on this choice (Fig. 1).While WGS approach can capture all possible mutations,WES or targeted gene panel sequencing (targeted NGS)are cost-effective approaches for capturing phenotypealtering mutations (2, 3). With unlimited resources andtime, WGS is a clear winner as it allows you to interrogateSNVs, insertions or deletions (indels), structural variants(SVs) and copy number variants (CNVs) in both the1% part of the genome that encodes protein sequencesand the 99% of remaining non-coding regions. WES isfocused on the detection of SNVs and indels in proteincoding genes and on other functional elements such asmicroRNA sequences; consequently, it omits regulatoryregions such as promoters and enhancers. Although costsvary depending on the sequence capture solution, WEScan be an order of magnitude less expensive than WGS toachieve an approximately equivalent breadth of coverageof protein-coding exons. These reduced costs offer thepotential to greatly increase sample numbers, which isa key factor for many studies and clinical applications.On the other hand, targeted NGS represents the cheapersolution adopted in a great number of laboratoriesallowing the optimization of different features ofmolecular diagnostics workflow (reduced costs for librarypreparation steps and sequencing run, adaptable todifferent kind of samples, increased number of samplesprocessed in each run and reduction of time needed forNGS in endocrine diseases179:3R112clinical counselling). In Table 1, we report the potentialadvantages and limitations of those approaches (1, 2, 3,4, 5, 6).Study design for differentsequencing strategyThe choice of the sequencing strategy should be determinedby the aim, the underlying biological hypothesis and sizeof the study. The targeted NGS, WES or WGS support thedifferent testing strategies, going from the sequencingof candidate genes selected for a particular disorder orphenotype up to an unsupervised sequencing of all genesin the genome.Studies of population genomics are mainly orientedon WGS strategy, they benefit from a trade-off betweensample numbers and sequencing depth, in which manygenomes are sequenced at low depth and variants aresimultaneously called across all samples. Variant calls onindividual low-depth genomes have a high false-positiverate, but this problem is overcome by combining resultsacross a high number of samples (7, 8, 9).When the aim of the study is to identify new genesinvolved in a particular disease, WES usually appears as themost convenient choice since it is considerably cheaperthan WGS and allows the sequencing of a consistentnumber of subjects with a good detection quality of SNVs,SVs and indels, but not for CNVs (10, 11).The analysis of gene panels isolated by targeted capturerepresents a valid alternative for diagnostic applications,when the lab should test only causative genes and givea response in a short time. The restricted targetingrepresents a relevant advantage as it reduces the possibilityof incidental findings thus facilitating interpretation ofgenetic results and allowing higher coverage at lower costthan genome/exome-wide approaches (Table 1).Notwithstanding differences among protocols andstudy design, the analytical approach is similar amongstrategies: sequencing step is followed by bioinformaticsanalysis and filtering of genetic variability aimed toselect genetic variants that are considered clinicallyrelevant. In such context, WES and WGS offer theadvantage of expanding the search space to consideradditional genes or genomic regions that have not beenpreviously identified but that may potentially explain anindividual’s complex or peculiar clinical presentation.However, a single exome can produce approximately30 K variants, while more than 3 M variants can befound in a single genome. Both in diagnostic and inwww.eje-online.orgDownloaded from Bioscientifica.com at 05/17/2022 12:43:34AMvia free access

L Persani and othersNGS in endocrine diseases179:3R113European Journal of EndocrinologyReviewFigure 1Schematic illustration of the algorithm for the choice of the most suitable next-generation sequencing (NGS) approach. TargetedNGS is generally more suitable for clinical applications. The whole exome or whole genome sequencing (WES or WGS) approacheshave mainly research applications, but positive results should expand the targeted NGS approaches. WES can also have a clinicalapplication for the diagnosis of particular heritable conditions that are not clinically defined. VUS, variants of uncertainsignificance.research areas, the bioinformatics task is fundamentalbut is therefore often long and challenging. Apart fromthe sequencing strategy, the most important challengetoday is represented by the analysis and classificationof genomic variants that have not been previouslyreported in the medical literature or in public databases(see below). The assessment of the variant pathogenicimpact should follow accepted guidelines for variantclassification (1). The classification of genetic variabilityand the determination of its role on human phenotyperepresent one of the most important challenges forclinicians, geneticists and researchers.NGS application for rare or commoncomplex diseasesNGS can be applied to study both rare diseases andcomplex diseases. Different examples demonstrate thepower of NGS in identifying causal variants for raremonogenic diseases even with very small sample size(12, 13, 14).However, not all rare conditions reflect thescenario of typical monogenic diseases with Mendelianinheritance. Indeed, some rare conditions includesimilar manifestations, and the same manifestation canwww.eje-online.orgDownloaded from Bioscientifica.com at 05/17/2022 12:43:34AMvia free access

ReviewL Persani and othersNGS in endocrine diseases179:3R114Table 1 Principal applications, advantages and limitations of the three different next-generation sequencing (NGS) strategies.NGS strategyPrincipal applicationWhole genomesequencing (WGS)Studies of population Identification of SNVs, indels, SV andgenomicsCNVs in coding and non-coding regions PCR amplification not required, reducingthe potential of GC bias More consistent and uniform coveragecompared to WES or Targeted NGS A lower average read depth is requiredto achieve the same coverage as WES No impact by sequencing read length Performance unaffected by capturing oramplification procedures Specific protocols of enrichment are notrequired Complete information on the geneticvariability of each sampleIdentification of new Identification of SNVs, indels, SV andcausative genesCNVs in coding regions Reduction of the cost in comparison withWGS Reduced number of identifiedgenetic variants with a reduction ofresources needed for their storage andinterpretation Increased number of samples analysed More suitable than WGS for clinicalapplicationsAnalysis of known Most suitable for clinical applicationscausative genes Higher coverage and sequencing depththan WES Customizable for different samples types,e.g. formalin-fixed paraffin-embeddedtissues, cell free or circulating tumouralDNA, degraded samples Highest number of samples Reduced computational and storageresources Lower testing costs than WGS and WES Genetic variability is determined only forselected genes thus reducing the risk ofincidental findingsEuropean Journal of EndocrinologyWhole exomesequencing (WES)Targeted NGSAdvantagesbe induced by different mechanisms. In such context,the identification of causal variants generally requireslarger sample sizes than classic monogenic diseasesor affected families with multiple generations (15). Incomplex diseases, there is an extreme heterogeneityin both the clinical spectrum and underlyingmechanisms: individuals with similar phenotypesmay involve different causal variants from the samegene or multiple variants in different genes actingwithin the disease pathway(s). Conversely, patientswith the same causal genetic factor may manifest avariable phenotype due to incomplete penetrance, asa consequence of the interaction with other genetic,epigenetic or environmental modifying factors. Inthis field, the characterization of genetic aetiology canbecome difficult.Limitations High cost Low number of samples Huge amount of genetic variantsidentified Greater effort in the datainterpretation Limited applications in routinediagnostics Assessment of genetic variabilityonly in exons Heterogeneous coverageinfluenced by library preparationprocedures High risk of genetic incidentalfindings Genetic analysis restricted toselected regions Variable and heterogeneouscoverage Problematic design of probes Biases of library preparation:false-positive variants due to PCRduplicates or false negative resultsdue to allelic drop out Difficult identification of CNVsA large survey of human genetic variation (16) showsthat rare variants represent the great part ( 70–80%) ofgenetic variability, and the UK10K project identifiedmore than 42 M SNVs over 3781 subjects (SNVs, 34.2 Mrare and 2.2 M low frequency). Statistical geneticsconsiderations of rare variant association analysis havebeen the focus of intensive method development over thelast few years in the field of complex traits studies. Evenif there is a substantial contribution from rare variants,it remains challenging to detect rare variant effects dueto low statistical power. Deep WGS of large numbersof individuals would represent the most informativestrategy for association studies of complex traits anddiseases. However, large-scale WGS is generally unfeasiblein the field of classical epidemiological designs, suchas case–control and cohort studies, because of the highwww.eje-online.orgDownloaded from Bioscientifica.com at 05/17/2022 12:43:34AMvia free access

European Journal of EndocrinologyReviewL Persani and otherscost. Several less costly sequencing strategies have beenproposed and used in common traits studies, such aslow-depth WGS, WES and targeted NGS. Many statisticalmethods have been then proposed to increase the signalor reduce the noise in testing variant-disease associationusing sequencing data. These statistical methods canbe classified into three categories: (i) the BURDEN test(17, 18), (ii) the sequence kernel association test (SKAT)(19) and (iii) the P values combination methods (20).Once a gene/region emerged as significantly associated,the next important step is to identify rare causal variantswithin these regions/genes. Identifying a small numberof rare causal variants that contribute to complexdiseases has become a major focus of investigation. Eachof the above gene-based methods reports a P value forthe association of multiple rare variants and a specificphenotypic trait. However, the following identification ofa small proportion of truly causal variants is an even moredifficult challenge and often need other approaches suchas in vitro or in vivo functional studies.Sequencing errors and confirmationof resultsMore than 200 000 genomes and an even higher numberof exomes have been sequenced to date. It is still widelyestablished that variants found using NGS should bevalidated with the current ‘gold standard’ for DNAsequencing, Sanger sequencing (21), though severalreports suggest that NGS results are at least as accurateor in some cases more accurate than Sanger sequencing(22, 23). Massively parallel sequencing technologies haverevolutionized medical genetics, however, also NGS isprone to both negative and positive results. Problems maybe generated during library preparation procedure, PCRartefacts for example, can introduce false-positive resultsin capturing-based libraries, while amplicon-based librariesare prone to allelic dropout problems due to presenceof variation in the sequence that produce the selectiveamplification of a single allele. Errors can also be introducedduring the bioinformatics analysis, extended insertions ordeletions for example can be missed. To date, there are novalidated procedures to detect sequencing errors.NGS for discovery of novelpathogenic mechanismsConventionally, the diagnostic approach to endocrinediseases was based on physical findings, biochemicalNGS in endocrine diseases179:3R115testing and imaging analysis, followed, secondly bymolecular analysis. Until recently, DNA samples wereanalysed following a phenotype-driven strategy andusing Sanger sequencing method of the coding regions ofone candidate gene at a time, an operation that requiresseveral weeks for a response.The scenario has dramatically changed with theintroduction of NGS. The outcomes, related consequencesand risks are summarized in Table 2. With the NGSintroduction, the time needed to systematically sequencea set of candidate genes has decreased from several weeksto few days. This finally has progressively brought downthe costs for a massive parallel high-throughput DNAsequencing and a comprehensive genetic diagnosis.However, this advancement becomes possible with theintroduction of new key-personnel for the design of NGSexperiment and bioinformatics data analysis.An increasing number of scientific reports ( 600)using NGS for endocrine disease investigations werepublished in peer-reviewed journals since 2009 (Fig. 2).Here, we selected some examples showing how NGSappeared successful in identifying new causative genes orpathogenic mechanisms.One of these studies is on familial central precociouspuberty (CPP) (24). The Brazilian authors performeda WES in 40 members of 15 families affected by CPP ofunknown aetiology. NGS analysis identified MKRN3 geneas a potential causative candidate gene for the disease. Theimportant role of MKRN3 in human puberty initiation wasreinforced by large genome-wide studies involving womenof European descent from 57 studies (25). Subsequently,several studies confirmed that mutations in the paternallyexpressed imprinted MKRN3 gene constitute a majorgenetic cause of heritable or apparently sporadic CPP(26, 27). Very recently, a genomic defect in DLK1, a geneencoding a ligand of the Notch receptors, was discoveredby NGS as being also associated with isolated familial CPP(28). Interestingly, MKRN3 and DLK1 are both paternallyexpressed imprinted genes. These findings suggest a roleof genomic imprinting in regulating the timing of humanpuberty.Importantly, MKRN3 falls in the locus on chromosome15 that is known to be associated with andneurological dysfunctions associated with severe obesitydue to a complete resistance to satiety signals), whereasDLK1 had been previously implicated in Temple syndrome(including precocious puberty with intrauterine growthretardation, postnatal short stature, hypotonia, smallhands and mild facial dysmorphisms) (28). Therefore,www.eje-online.orgDownloaded from Bioscientifica.com at 05/17/2022 12:43:34AMvia free access

ReviewL Persani and othersNGS in endocrine diseases179:3R116Table 2 Novel scenarios opened by the NGS approach offering massive parallel high-throughput DNA sequencing for researchor diagnosis of endocrine conditions.NGS aftereffects (ref.)Comments Novel personnel requirements bioinformatics forexperiment planning and interpretation of results Potential explanation of patients previously classified asphenocopiesIdentification of novel causative genes (24, 28, 40, 46, 71, 76) Expanded understanding of endocrine conditions Novel pathogenic mechanismsGenetic classification of endocrine conditions (44, 50, 51, 52, Precision medicine targeted therapies and management,53, 54, 57, 61, 63, 64, 67, 68, 72, 75)and accurate prognosisIdentification of multigenic involvement (30, 31, 32, 33, 34, Novel pathogenic mechanisms38, 41, 42) Possible explanation for the variable expressivity andpenetrance of certain candidate gene variationsGenetic heterogeneity of one condition (34, 36, 38, 42, 43, 44, Expanded understanding of endocrine conditions57, 58, 59, 75) Novel pathogenic mechanisms Allelic quantification more defined prognosis for endocrinetumoursOne gene associated to multiple clinical conditions Role of gene modifiers(26, 28, 29, 31, 37, 38, 40) Expanded understanding of gene function Novel pathogenic mechanisms Challenging genetic counsellingDefined frequency of gene variants (46, 57, 61, 62, 63, 64, 67, Classification of gene variants68, 77) Improved genetic counsellingUnexpected variants in genes unrelated to the investigated Ethical issuescondition (incidental findings) (81) Challenging genetic counselling Adequate information for patient’s consentIdentification of variants of unknown significance (78, 79, 80) Adequate bioinformatic and clinical classification Challenging genetic counsellingEuropean Journal of EndocrinologyReduced time and costs and high accuracy for geneticdiagnosis (1, 2, 3, 39, 78, 79)one additional consequence of the NGS approach is theunexpected discovery of the association between a singlegene and two apparently distinct disorders. Anotherexample of this kind concerns the JAG1 gene that encodesanother ligand of the Notch receptors and was originallylinked to Alagille syndrome (a disorder characterized bycholestasis, heart malformations, together with eye, facialand skeletal abnormalities, and by an extremely variableexpressivity and penetrance of the heterozygous JAG1mutations) (29). By an NGS approach, we observed thatmonoallelic JAG1 variants are frequently associated withrare variants affecting thyroid specific genes in severalnewborns affected with congenital hypothyroidism (CH),another disorder characterized by variable expressivityand penetrance of the candidate gene defects (30). Arole for JAG1 in thyroid development was supported byin vivo studies in the zebrafish model, and it is thereforepossible that JAG1 minor alleles could contribute toCH pathogenesis by acting as genetic modifiers andamplifying the partial loss of function associated withmonoallelic defects in thyroid-specific genes (31).Our NGS data obtained in a large Italian cohort ofCH patients and in an ethnicity-matched population areconsistent with an oligogenic model of CH pathogenesis(30). We found several variants of 11 genes with arelevant role in thyroid morphogenesis or function thathave a rare or low frequency in the general population,but a significant enrichment in the CH population.When expressed alone in the first-degree relatives of CHpatients, these rare variants were associated with minorFigure 2Timeline of articles in PubMed with the key words: Nextgeneration sequencing AND endocrine disease (updatedMarch 2018).www.eje-online.orgDownloaded from Bioscientifica.com at 05/17/2022 12:43:34AMvia free access

European Journal of EndocrinologyReviewL Persani and othersthyroid defects, whereas the variable combinations of 2minor alleles were unexpectedly found in about 25% ofCH patients. Other groups obtained similar NGS findingsin smaller CH cohorts (32, 33). Such an oligogenic originof CH provides a suitable explanation for the frequentsporadic appearance of CH in the population. In all theseNGS studies targeted to a panel of known CH candidategenes, the frequency of variations found by theirsystematic and unsupervised analyses was unexpectedlyhigher that the rate of positive findings in previousstudies performed by Sanger in phenotypically selectedCH cohorts (5–20% depending upon the investigatedphenotype). This might be explained both by an increasedsensitivity of NGS protocols in the genetic variationdetection as well as by the involvement of morphogeneticor functional genes independently of the observed CHphenotype. Noteworthy, the number of cases remainingunexplained after these targeted NGS analyses falls downbelow 40% thus representing a significant improvementfor the genetic counselling in clinical practice (34).In other cases, NGS allowed to link one gene to twoendocrine conditions that were previously consideredcompletely distinct. Izumi et al. identified by NGS a SOX3polyalanine deletion in a patient with normosmic isolatedhypogonadotropic hypogonadism (IHH) (35). This findingexpanded the phenotypic spectrum of SOX3 polyalaninedeletion to include IHH without other pituitary hormonedeficiencies or mental retardation (36). Moreover, theydetected a WDR11 splice-site mutation in a patientwith multiple pituitary hormone deficiency (MPHD)(35). These data indicate that WDR11, a gene known tobe involved in normosmic IHH, can also cause MPHDs.Similar results had been previously reported for otherIHH and Kallmann syndrome (KS: IHH with anosmia)genes, such as PROKR2 or FGF8 and FGFR1 that have beendescribed in MPHDs also (37). The clinical presentationof heterozygous defects in candidate genes for IHH/KS is known to be highly variable among and withinthe same families which, in combination with the 30known candidate genes (38), makes the phenotype-drivengenetic analyses very complicated and expensive, as wellas disturbing and time consuming for the patients (39).The panel of candidate genes was further expanded by anNGS study conducted on 261 genes (known to be involvedin hypothalamic, pituitary, and/or olfactory pathways, orsuggested by chromosome rearrangements) that identified18 new potential candidate genes for IHH/KS (40). Insuch heterogeneous-related conditions (IHH, KS, MPHDs)associated with a high number of candidate genes, theapplication of NGS protocols is now unavoidable. IndeedNGS in endocrine diseases179:3R117the systematic NGS analyses will help to define the exactfrequency of involvement of single genes and the clinicalimpact of oligogenic involvement (41).This picture is very similar to that now seen in differentclinical settings, such as the primary ovarian insufficiency(POI) or the disorders of sex development (DSD) (42, 43).The genetic diagnosis in POI patients may become usefulfor the preservation of fertility in the affected families orin the identification of patients with higher chances toobtain a fertilizable egg or at higher risk of extra-ovariandefects (44). The Vilain Lab conducted two studies onDSD patients (44, 45). In the first, they proposed a novelprocedure, reversing the order of the diagnostic endocrinesteps and starting from the genetic analysis. By thisapproach, they claim the possibility to eliminate nonindicated clinical tests, sparing the patient unnecessarystress and saving healthcare system’s resources. Theydesigned a targeted panel sequencing of 35 DSD genesthat revealed genetic defects in two out of seven patientsnot previously diagnosed and confirmed the diagnosis inanother seven patients with known genetic causes (45).In the second study, they performed a WES expandingthe list of genes to analyse 64 candidates. They reacheda diagnostic yield of 35% in 40 patients with 46,XY DSDwho had not previously received a genetic diagnosis (46).The application of NGS recently expanded the viewon the mechanisms underlying tumour formation.Until recently, activating mutations in the TSH receptor(TSHR) and in GNAS represented the principal causeof autonomous thyroid adenomas (ATAs) and wereconsidered oncogenes sufficient to induce autonomousfunction and growth. In a study conducted on severalATAs by WES, we found that a relevant fraction ofATAs carry, beyond the well-known TSHR and GNASvariants, also a recurrent hot-spot mutation in EZH1, akey gene involved in the epigenetic regulation of celldifferentiation and proliferation. Interestingly, the EZH1variant was found to be associated with the TSHR or GNASvariants suggesting a 2-hit model for the pathogenesis ofthese benign tumours, whereby constitutive activation ofthe cAMP pathway and EZH1 mutations may cooperate toinduce the hyperproliferation of thyroid cells (47).NGS in endocrine tumours: role indifferential diagnosis and predictionof outcomeIn the last 10 years, NGS technology led to a betterknowledge in all human cancers, including endocrine,www.eje-online.orgDownloaded from Bioscientifica.com at 05/17/2022 12:43:34AMvia free access

European Journal of EndocrinologyReviewL Persani and otherssporadic and familial tumours. More importantly than inother clinical conditions, these new genetic knowledgewill become more and more useful for clinicians in thepersonalized management of patients.Cancer is a heterogeneous disease harbouring differentsubclonal cell populations that can be discriminated bytheir DNA mutations. The genome study of a cancer canhelp to better identify its heterogeneity and to supportthe clinicians in the choice of a more effective treatmentfor each patient. For this reason, NGS applicationsare becoming an integral part of the clinical routinediagnostics in different endocrine cancers.In thyroid tumours, NGS represents the gold standardtechnique for the pre-surgical molecular diagnosis ofthyroid nodules and for the molecular profiling ofthyroid carcinoma. Fine-needle aspiration cytology is thestandard pre-operative tool for thyroid nodule diagnosis,however, up to 25–30% of the samples are classified asindeterminate (48). In these cases, the guidelines of theAmerican Thyroid Association recommend moleculartesting in order to better identify malignant samples andto plan patient’s management (49). In order to improvediagnosis and optimize the management of thyroidnodules with indeterminate cytological diagnosis,Nikiforov et al. developed a targeted NGS test, ThyroSeqv2, which includes analyses of point mutations, genefusions and abnormal gene expression in 56 thyroidrelated genes (50). Subsequently, various groups validatedthe test (51, 52) that was very recently expanded to include112 genes, the ThyroSeq v3 (53). This panel now includesalso several genes whose role in thyroid carcinogenesisremains elusive (e.g. TSHR), thus making indefinite theinterpretation of several potential variations. However,the integration of the genetic and cytology results iscruci

genomes are sequenced at low depth and variants are simultaneously called across all samples. Variant calls on individual low-depth genomes have a high false-positive rate, but this problem is overcome by combining results across a high number of samples (7, 8, 9). When the aim of the study is to identify new genes

Related Documents:

Genetics – A Continuity of Life – Daniel Fairbanks, Ralph Anderson. Concepts of Genetics – Klug and Cummings. Principles of Genetics – Hartt and Jones. GN 5 B 07 : CORE COURSE VII Medical Genetics Total – 54 hrs Unit- 1: Principles of Human Genetics (2 hrs) History, Origin of medical genetics, classification of genetic disease .

The endocrinology and evolution of tropical social wasps: from casteless groups to high societies Chapter 1 (pp. 7-44) A brief review of social insect endocrinology with an emphasis on the Hymenoptera Chapter 2 (pp. 45-72) The endocrinology and behavior of Zethus miniatus Chapter 3 (pp.73-125) The endocrinology of Polybia micans

Common Endocrinology Conditions Many children/adolescents struggle with one or more of these four common endocrinology issues: abnormal menstrual patterns, abnormal thyroid, short stature and/or pediatric obesity . To assist with patient diagnosis and referral, the Children's Mercy Endocrinology team created these recommendations. Winter 2021

GENETICS: INTRODUCTION TO GENETIC COUNSELING . In many genetics clinics, genetic counselors and/or genetic nurses work with MD clinical geneticists. Although each clinic is unique, often counselors and nurses provide the bulk of information gathering, risk assessment, and counseling. MD cli

Pediatric Endocrinology Referral Guidelines Thank you for referring your patients to our division. We are striving to provide the best possible experience . The Division of Pediatric Endocrinology 'Specially for Children Dell Children's Medical Center of Central Texas. For appointments, please call the Endocrinology Center at (512) 628 .

The division runs two inpatient services, General Endocrine and an Inpatient . Pediatric Endocrinology. Reproductive Endocrinology. . Endocrinology Fellowship Program. University of Mississippi Medical Center . 2500 N. State St. Jackson, Ms 39216 (601)-984-5525.

Chief, Division of Endocrinology and Metabolism Affiliated with the University of Pittsburgh School of Medicine, UPMC is proud to be nationally ranked by U.S. News & World Report for excellence in endocrinology. FALL 2020 DIVISION OF ENDOCRINOLOGY AND METABOLISM Inside This Edition 2 Intestinal Lipoproteins in Normal Metabolism and Disease

division of reproductive endocrinology and infertilit y. paula amato, m.d. professor of obste tric s and gynecology division of reproductive endocrinology and infertilit y. diana wu, m.d. associate professor of obste tric s and gynecology division of reproductive endocrinology and infertilit y. thomas o'leary, ph.d. director, fertility laboratory