Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Hum Mol Genet ; 29(13): 2109-2123, 2020 08 03.
Article En | MEDLINE | ID: mdl-32186706

Cobalamin C (cblC) deficiency, the most common inborn error of intracellular cobalamin metabolism, is caused by mutations in MMACHC, a gene responsible for the processing and intracellular trafficking of vitamin B12. This recessive disorder is characterized by a failure to metabolize cobalamin into adenosyl- and methylcobalamin, which results in the biochemical perturbations of methylmalonic acidemia, hyperhomocysteinemia and hypomethioninemia caused by the impaired activity of the downstream enzymes, methylmalonyl-CoA mutase and methionine synthase. Cobalamin C deficiency can be accompanied by a wide spectrum of clinical manifestations, including progressive blindness, and, in mice, manifests with very early embryonic lethality. Because zebrafish harbor a full complement of cobalamin metabolic enzymes, we used genome editing to study the loss of mmachc function and to develop the first viable animal model of cblC deficiency. mmachc mutants survived the embryonic period but perished in early juvenile life. The mutants displayed the metabolic and clinical features of cblC deficiency including methylmalonic acidemia, severe growth retardation and lethality. Morphologic and metabolic parameters improved when the mutants were raised in water supplemented with small molecules used to treat patients, including hydroxocobalamin, methylcobalamin, methionine and betaine. Furthermore, mmachc mutants bred to express rod and/or cone fluorescent reporters, manifested a retinopathy and thin optic nerves (ON). Expression analysis using whole eye mRNA revealed the dysregulation of genes involved in phototransduction and cholesterol metabolism. Zebrafish with mmachc deficiency recapitulate the several of the phenotypic and biochemical features of the human disorder, including ocular pathology, and show a response to established treatments.


Carrier Proteins/genetics , Morphogenesis/genetics , Vitamin B 12 Deficiency/genetics , Vitamin B 12/genetics , Zebrafish Proteins/genetics , Animals , Homocystinuria/genetics , Homocystinuria/pathology , Humans , Mice , Mutation/genetics , Optic Nerve/growth & development , Optic Nerve/pathology , Oxidoreductases/genetics , Retina/growth & development , Retina/metabolism , Vitamin B 12/analogs & derivatives , Vitamin B 12/metabolism , Vitamin B 12 Deficiency/metabolism , Vitamin B 12 Deficiency/pathology , Zebrafish/genetics , Zebrafish/growth & development
2.
PLoS Genet ; 14(12): e1007821, 2018 12.
Article En | MEDLINE | ID: mdl-30540754

Fanconi Anemia (FA) is a genomic instability syndrome resulting in aplastic anemia, developmental abnormalities, and predisposition to hematological and other solid organ malignancies. Mutations in genes that encode proteins of the FA pathway fail to orchestrate the repair of DNA damage caused by DNA interstrand crosslinks. Zebrafish harbor homologs for nearly all known FA genes. We used multiplexed CRISPR/Cas9-mediated mutagenesis to generate loss-of-function mutants for 17 FA genes: fanca, fancb, fancc, fancd1/brca2, fancd2, fance, fancf, fancg, fanci, fancj/brip1, fancl, fancm, fancn/palb2, fanco/rad51c, fancp/slx4, fancq/ercc4, fanct/ube2t, and two genes encoding FA-associated proteins: faap100 and faap24. We selected two indel mutations predicted to cause premature truncations for all but two of the genes, and a total of 36 mutant lines were generated for 19 genes. Generating two independent mutant lines for each gene was important to validate their phenotypic consequences. RT-PCR from homozygous mutant fish confirmed the presence of transcripts with indels in all genes. Interestingly, 4 of the indel mutations led to aberrant splicing, which may produce a different protein than predicted from the genomic sequence. Analysis of RNA is thus critical in proper evaluation of the consequences of the mutations introduced in zebrafish genome. We used fluorescent reporter assay, and western blots to confirm loss-of-function for several mutants. Additionally, we developed a DEB treatment assay by evaluating morphological changes in embryos and confirmed that homozygous mutants from all the FA genes that could be tested (11/17), displayed hypersensitivity and thus were indeed null alleles. Our multiplexing strategy helped us to evaluate 11 multiple gene knockout combinations without additional breeding. Homozygous zebrafish for all 19 single and 11 multi-gene knockouts were adult viable, indicating FA genes in zebrafish are generally not essential for early development. None of the mutant fish displayed gross developmental abnormalities except for fancp-/- fish, which were significantly smaller in length than their wildtype clutch mates. Complete female-to-male sex reversal was observed in knockouts for 12/17 FA genes, while partial sex reversal was seen for the other five gene knockouts. All adult females were fertile, and among the adult males, all were fertile except for the fancd1 mutants and one of the fancj mutants. We report here generation and characterization of zebrafish knockout mutants for 17 FA disease-causing genes, providing an integral resource for understanding the pathophysiology associated with the disrupted FA pathway.


Fanconi Anemia/genetics , Zebrafish/genetics , Animals , CRISPR-Cas Systems , DNA Damage , Fanconi Anemia/physiopathology , Female , Fertility/genetics , Fertility/physiology , Frameshift Mutation , Gene Knockout Techniques , Humans , Male , Phenotype , RNA Splicing/genetics , Sex Determination Processes/genetics , Sex Determination Processes/physiology , Sexual Development/genetics , Sexual Development/physiology , Zebrafish/growth & development , Zebrafish/physiology , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
3.
NPJ Regen Med ; 3: 11, 2018.
Article En | MEDLINE | ID: mdl-29872546

Regenerative medicine holds great promise for both degenerative diseases and traumatic tissue injury which represent significant challenges to the health care system. Hearing loss, which affects hundreds of millions of people worldwide, is caused primarily by a permanent loss of the mechanosensory receptors of the inner ear known as hair cells. This failure to regenerate hair cells after loss is limited to mammals, while all other non-mammalian vertebrates tested were able to completely regenerate these mechanosensory receptors after injury. To understand the mechanism of hair cell regeneration and its association with regeneration of other tissues, we performed a guided mutagenesis screen using zebrafish lateral line hair cells as a screening platform to identify genes that are essential for hair cell regeneration, and further investigated how genes essential for hair cell regeneration were involved in the regeneration of other tissues. We created genetic mutations either by retroviral insertion or CRISPR/Cas9 approaches, and developed a high-throughput screening pipeline for analyzing hair cell development and regeneration. We screened 254 gene mutations and identified 7 genes specifically affecting hair cell regeneration. These hair cell regeneration genes fell into distinct and somewhat surprising functional categories. By examining the regeneration of caudal fin and liver, we found these hair cell regeneration genes often also affected other types of tissue regeneration. Therefore, our results demonstrate guided screening is an effective approach to discover regeneration candidates, and hair cell regeneration is associated with other tissue regeneration.

4.
Nat Protoc ; 11(12): 2357-2375, 2016 Dec.
Article En | MEDLINE | ID: mdl-27809318

The zebrafish is a popular model organism for studying development and disease, and genetically modified zebrafish provide an essential tool for functional genomic studies. Numerous publications have demonstrated the efficacy of gene targeting in zebrafish using CRISPR/Cas9, and they have included descriptions of a variety of tools and methods for guide RNA synthesis and mutant identification. However, most of the published techniques are not readily scalable to increase throughput. We recently described a CRISPR/Cas9-based high-throughput mutagenesis and phenotyping pipeline in zebrafish. Here, we present a complete workflow for this pipeline, including target selection; cloning-free single-guide RNA (sgRNA) synthesis; microinjection; validation of the target-specific activity of the sgRNAs; founder screening to identify germline-transmitting mutations by fluorescence PCR; determination of the exact lesion by Sanger or next-generation sequencing (including software for analysis); and genotyping in the F1 or subsequent generations. Using these methods, sgRNAs can be evaluated in 3 d, zebrafish germline-transmitting mutations can be identified within 3 months and stable lines can be established within 6 months. Realistically, two researchers can target tens to hundreds of genes per year using this protocol.


CRISPR-Cas Systems/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Mutagenesis , Zebrafish/genetics , Animals
5.
Sci Rep ; 6: 29946, 2016 07 18.
Article En | MEDLINE | ID: mdl-27425195

Phosphoribosyl pyrophosphate synthetase-1 (PRPS1) is a key enzyme in nucleotide biosynthesis, and mutations in PRPS1 are found in several human diseases including nonsyndromic sensorineural deafness, Charcot-Marie-Tooth disease-5, and Arts Syndrome. We utilized zebrafish as a model to confirm that mutations in PRPS1 result in phenotypic deficiencies in zebrafish similar to those in the associated human diseases. We found two paralogs in zebrafish, prps1a and prps1b and characterized each paralogous mutant individually as well as the double mutant fish. Zebrafish prps1a mutants and prps1a;prps1b double mutants showed similar morphological phenotypes with increasingly severe phenotypes as the number of mutant alleles increased. Phenotypes included smaller eyes and reduced hair cell numbers, consistent with the optic atrophy and hearing impairment observed in human patients. The double mutant also showed abnormal development of primary motor neurons, hair cell innervation, and reduced leukocytes, consistent with the neuropathy and recurrent infection of the human patients possessing the most severe reductions of PRPS1 activity. Further analyses indicated the phenotypes were associated with a prolonged cell cycle likely resulting from reduced nucleotide synthesis and energy production in the mutant embryos. We further demonstrated the phenotypes were caused by delays in the tissues most highly expressing the prps1 genes.


Ribose-Phosphate Pyrophosphokinase/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Brain/embryology , Brain/metabolism , Ear, Inner/embryology , Ear, Inner/innervation , Ear, Inner/metabolism , Embryo, Nonmammalian/metabolism , Eye/metabolism , Eye/pathology , Gene Expression Regulation, Developmental , Hematopoiesis , Humans , Leukocytes/metabolism , Models, Biological , Motor Neurons/metabolism , Mutation/genetics , Phenotype , Pigmentation/genetics , Ribose-Phosphate Pyrophosphokinase/genetics , S-Adenosylmethionine/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Hum Mutat ; 37(5): 465-8, 2016 May.
Article En | MEDLINE | ID: mdl-26841305

Fanconi anemia (FA) is a rare inherited disorder caused by pathogenic variants in one of 19 FANC genes. FA patients display congenital abnormalities, and develop bone marrow failure, and cancer susceptibility. We identified homozygous mutations in four FA patients and, in each case, only one parent carried the obligate mutant allele. FANCA and FANCP/SLX4 genes, both located on chromosome 16, were the affected recessive FA genes in three and one family respectively. Genotyping with short tandem repeat markers and SNP arrays revealed uniparental disomy (UPD) of the entire mutation-carrying chromosome 16 in all four patients. One FANCA patient had paternal UPD, whereas FA in the other three patients resulted from maternal UPD. These are the first reported cases of UPD as a cause of FA. UPD indicates a reduced risk of having another child with FA in the family and has implications in prenatal diagnosis.


Chromosomes, Human, Pair 16/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia/genetics , Recombinases/genetics , Uniparental Disomy/genetics , Adult , Child, Preschool , Female , Genes, Recessive , Homozygote , Humans , Male , Mutation , Pedigree , Polymorphism, Single Nucleotide , Young Adult
7.
J Exp Med ; 212(8): 1185-202, 2015 Jul 27.
Article En | MEDLINE | ID: mdl-26150473

Adenylate kinases (AKs) are phosphotransferases that regulate the cellular adenine nucleotide composition and play a critical role in the energy homeostasis of all tissues. The AK2 isoenzyme is expressed in the mitochondrial intermembrane space and is mutated in reticular dysgenesis (RD), a rare form of severe combined immunodeficiency (SCID) in humans. RD is characterized by a maturation arrest in the myeloid and lymphoid lineages, leading to early onset, recurrent, and overwhelming infections. To gain insight into the pathophysiology of RD, we studied the effects of AK2 deficiency using the zebrafish model and induced pluripotent stem cells (iPSCs) derived from fibroblasts of an RD patient. In zebrafish, Ak2 deficiency affected hematopoietic stem and progenitor cell (HSPC) development with increased oxidative stress and apoptosis. AK2-deficient iPSCs recapitulated the characteristic myeloid maturation arrest at the promyelocyte stage and demonstrated an increased AMP/ADP ratio, indicative of an energy-depleted adenine nucleotide profile. Antioxidant treatment rescued the hematopoietic phenotypes in vivo in ak2 mutant zebrafish and restored differentiation of AK2-deficient iPSCs into mature granulocytes. Our results link hematopoietic cell fate in AK2 deficiency to cellular energy depletion and increased oxidative stress. This points to the potential use of antioxidants as a supportive therapeutic modality for patients with RD.


Adenylate Kinase/metabolism , Hematopoietic Stem Cells/physiology , Leukopenia/enzymology , Leukopenia/physiopathology , Oxidative Stress/physiology , Pluripotent Stem Cells/physiology , Severe Combined Immunodeficiency/enzymology , Severe Combined Immunodeficiency/physiopathology , Acridine Orange , Adenylate Kinase/deficiency , Animals , Antioxidants/pharmacology , Apoptosis/physiology , Azo Compounds , Base Sequence , Cell Differentiation/drug effects , Computational Biology , DNA Primers/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Molecular Sequence Data , Naphthalenes , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Zebrafish
8.
Genome Res ; 25(7): 1030-42, 2015 Jul.
Article En | MEDLINE | ID: mdl-26048245

The use of CRISPR/Cas9 as a genome-editing tool in various model organisms has radically changed targeted mutagenesis. Here, we present a high-throughput targeted mutagenesis pipeline using CRISPR/Cas9 technology in zebrafish that will make possible both saturation mutagenesis of the genome and large-scale phenotyping efforts. We describe a cloning-free single-guide RNA (sgRNA) synthesis, coupled with streamlined mutant identification methods utilizing fluorescent PCR and multiplexed, high-throughput sequencing. We report germline transmission data from 162 loci targeting 83 genes in the zebrafish genome, in which we obtained a 99% success rate for generating mutations and an average germline transmission rate of 28%. We verified 678 unique alleles from 58 genes by high-throughput sequencing. We demonstrate that our method can be used for efficient multiplexed gene targeting. We also demonstrate that phenotyping can be done in the F1 generation by inbreeding two injected founder fish, significantly reducing animal husbandry and time. This study compares germline transmission data from CRISPR/Cas9 with those of TALENs and ZFNs and shows that efficiency of CRISPR/Cas9 is sixfold more efficient than other techniques. We show that the majority of published "rules" for efficient sgRNA design do not effectively predict germline transmission rates in zebrafish, with the exception of a GG or GA dinucleotide genomic match at the 5' end of the sgRNA. Finally, we show that predicted off-target mutagenesis is of low concern for in vivo genetic studies.


CRISPR-Cas Systems , Gene Targeting , High-Throughput Screening Assays , Phenotype , Alleles , Animals , Gene Knockout Techniques , Gene Targeting/methods , Genome-Wide Association Study , Genomics , Germ Cells/immunology , Humans , Mutagenesis , Quantitative Trait Loci , RNA, Guide, Kinetoplastida/genetics , Sequence Deletion , Zebrafish
9.
Gastroenterology ; 149(1): 67-78, 2015 Jul.
Article En | MEDLINE | ID: mdl-25865046

BACKGROUND & AIMS: Small intestinal carcinoids are rare and difficult to diagnose and patients often present with advanced incurable disease. Although the disease occurs sporadically, there have been reports of family clusters. Hereditary small intestinal carcinoid has not been recognized and genetic factors have not been identified. We performed a genetic analysis of families with small intestinal carcinoids to establish a hereditary basis and find genes that might cause this cancer. METHODS: We performed a prospective study of 33 families with at least 2 cases of small intestinal carcinoids. Affected members were characterized clinically and asymptomatic relatives were screened and underwent exploratory laparotomy for suspected tumors. Disease-associated mutations were sought using linkage analysis, whole-exome sequencing, and copy number analyses of germline and tumor DNA collected from members of a single large family. We assessed expression of mutant protein, protein activity, and regulation of apoptosis and senescence in lymphoblasts derived from the cases. RESULTS: Familial and sporadic carcinoids are clinically indistinguishable except for the multiple synchronous primary tumors observed in most familial cases. Nearly 34% of asymptomatic relatives older than age 50 were found to have occult tumors; the tumors were cleared surgically from 87% of these individuals (20 of 23). Linkage analysis and whole-exome sequencing identified a germline 4-bp deletion in the gene inositol polyphosphate multikinase (IPMK), which truncates the protein. This mutation was detected in all 11 individuals with small intestinal carcinoids and in 17 of 35 family members whose carcinoid status was unknown. Mutant IPMK had reduced kinase activity and nuclear localization, compared with the full-length protein. This reduced activation of p53 and increased cell survival. CONCLUSIONS: We found that small intestinal carcinoids can occur as an inherited autosomal-dominant disease. The familial form is characterized by multiple synchronous primary tumors, which might account for 22%-35% of cases previously considered sporadic. Relatives of patients with familial carcinoids should be screened to detect curable early stage disease. IPMK haploinsufficiency promotes carcinoid tumorigenesis.


Carcinoid Tumor/genetics , Germ-Line Mutation , Intestinal Neoplasms/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoid Tumor/diagnosis , Carcinoid Tumor/pathology , Family , Female , Humans , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/pathology , Laparotomy , Male , Middle Aged , Pedigree , Prospective Studies , Young Adult
10.
Blood ; 124(1): 70-8, 2014 Jul 03.
Article En | MEDLINE | ID: mdl-24850758

CBFß and RUNX1 form a DNA-binding heterodimer and are both required for hematopoietic stem cell (HSC) generation in mice. However, the exact role of CBFß in the production of HSCs remains unclear. Here, we generated and characterized 2 zebrafish cbfb null mutants. The cbfb(-/-) embryos underwent primitive hematopoiesis and developed transient erythromyeloid progenitors, but they lacked definitive hematopoiesis. Unlike runx1 mutants, in which HSCs are not formed, nascent, runx1(+)/c-myb(+) HSCs were formed in cbfb(-/-) embryos. However, the nascent HSCs were not released from the aorta-gonad-mesonephros (AGM) region, as evidenced by the accumulation of runx1(+) cells in the AGM that could not enter circulation. Moreover, wild-type embryos treated with an inhibitor of RUNX1-CBFß interaction, Ro5-3335, phenocopied the hematopoietic defects in cbfb(-/-) mutants, rather than those in runx1(-/-) mutants. Finally, we found that cbfb was downstream of the Notch pathway during HSC development. Our data suggest that runx1 and cbfb are required at 2 different steps during early HSC development. CBFß is not required for nascent HSC emergence but is required for the release of HSCs from AGM into circulation. Our results also indicate that RUNX1 can drive the emergence of nascent HSCs in the AGM without its heterodimeric partner CBFß.


CCAAT-Binding Factor/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Zebrafish Proteins/genetics , Animals , CCAAT-Binding Factor/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Knockout Techniques , In Situ Hybridization , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish , Zebrafish Proteins/metabolism
11.
PLoS One ; 8(2): e57239, 2013.
Article En | MEDLINE | ID: mdl-23451191

Recently, it has been shown that targeted mutagenesis using zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) can be used to generate knockout zebrafish lines for analysis of their function and/or developing disease models. A number of different methods have been developed for the design and assembly of gene-specific ZFNs and TALENs, making them easily available to most zebrafish researchers. Regardless of the choice of targeting nuclease, the process of generating mutant fish is similar. It is a time-consuming and multi-step process that can benefit significantly from development of efficient high throughput methods. In this study, we used ZFNs assembled through either the CompoZr (Sigma-Aldrich) or the CoDA (context-dependent assembly) platforms to generate mutant zebrafish for nine genes. We report our improved high throughput methods for 1) evaluation of ZFNs activity by somatic lesion analysis using colony PCR, eliminating the need for plasmid DNA extractions from a large number of clones, and 2) a sensitive founder screening strategy using fluorescent PCR with PIG-tailed primers that eliminates the stutter bands and accurately identifies even single nucleotide insertions and deletions. Using these protocols, we have generated multiple mutant alleles for seven genes, five of which were targeted with CompoZr ZFNs and two with CoDA ZFNs. Our data also revealed that at least five-fold higher mRNA dose was required to achieve mutagenesis with CoDA ZFNs than with CompoZr ZFNs, and their somatic lesion frequency was lower (<5%) when compared to CopmoZr ZFNs (9-98%). This work provides high throughput protocols for efficient generation of zebrafish mutants using ZFNs and TALENs.


Gene Targeting , Mutagenesis , Zinc Fingers , Animals , Base Sequence , DNA Primers , Founder Effect , Germ Cells , Heterozygote , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Zebrafish
12.
Eur J Hum Genet ; 21(4): 437-43, 2013 Apr.
Article En | MEDLINE | ID: mdl-22948022

Prostate cancer (PrCa) is the most common male cancer in developed countries and the second most common cause of cancer death after lung cancer. We recently reported a genome-wide linkage scan in 69 Finnish hereditary PrCa (HPC) families, which replicated the HPC9 locus on 17q21-q22 and identified a locus on 2q37. The aim of this study was to identify and to detect other loci linked to HPC. Here we used ordered subset analysis (OSA), conditioned on nonparametric linkage to these loci to detect other loci linked to HPC in subsets of families, but not the overall sample. We analyzed the families based on their evidence for linkage to chromosome 2, chromosome 17 and a maximum score using the strongest evidence of linkage from either of the two loci. Significant linkage to a 5-cM linkage interval with a peak OSA nonparametric allele-sharing LOD score of 4.876 on Xq26.3-q27 (ΔLOD=3.193, empirical P=0.009) was observed in a subset of 41 families weakly linked to 2q37, overlapping the HPCX1 locus. Two peaks that were novel to the analysis combining linkage evidence from both primary loci were identified; 18q12.1-q12.2 (OSA LOD=2.541, ΔLOD=1.651, P=0.03) and 22q11.1-q11.21 (OSA LOD=2.395, ΔLOD=2.36, P=0.006), which is close to HPC6. Using OSA allows us to find additional loci linked to HPC in subsets of families, and underlines the complex genetic heterogeneity of HPC even in highly aggregated families.


Genetic Heterogeneity , Genetic Linkage , Prostatic Neoplasms/genetics , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 2/genetics , Female , Genetic Loci , Humans , Male , Pedigree
13.
Birth Defects Res A Clin Mol Teratol ; 94(11): 912-7, 2012 Nov.
Article En | MEDLINE | ID: mdl-22847929

BACKGROUND: Holoprosencephaly is the most frequent congenital malformation of the forebrain in humans. It is anatomically classified by the relative degree of abnormal formation and separation of the developing central nervous system. Mutations of ZIC2 are the second most common heterozygous variations detected in holoprosencephaly (HPE) patients. Mutations in most known HPE genes typically result in variable phenotypes that rage from classic alobar HPE to microforms represented by hypotelorism, solitary central maxillary incisor (SCMI), and cleft lip/palate, among others. Patients with HPE owing to ZIC2 mutations have recently been described by a distinct phenotype compared with mutations in other HPE causative genes. METHODS: We report the comparison of ZIC2 molecular findings by Sanger bidirectional DNA sequencing and ad hoc genotyping in a cohort of 105 Brazilian patients within the clinical spectrum of HPE, including classic and microform groups. RESULTS: We detected a total of five variants in the ZIC2 gene: a common histidine tract expansion c.716_718dup (p.His239dup), a rare c.1377_1391del_homozygous (p.Ala466_470del, or Ala 15 to 10 contraction), a novel intronic c.1239+18G>A variant, a novel frameshift c.1215dupC (p.Ser406Glnfs*11), and a c.1401_1406dup (p.Ala469_470dup, or alanine tract expansion to 17 residues). CONCLUSIONS: From these patients, only the latter two mutations found in classic HPE are likely to be medically significant. In contrast, variants detected in the microform group are not likely to be pathogenic. We show conclusively that the histidine tract expansion is a polymorphic alteration that demonstrates considerable differences in allele frequencies across different ethnic groups. Therefore, careful population studies of rare variants can improve genotype-phenotype correlations. Birth Defects Research (Part A) 2012.


Genetic Association Studies , Holoprosencephaly/genetics , Mutation , Nuclear Proteins/genetics , Transcription Factors/genetics , Adult , Alleles , Brazil/epidemiology , Case-Control Studies , Child , Child, Preschool , Female , Gene Frequency , Genotype , Heterozygote , Histidine/genetics , Holoprosencephaly/classification , Holoprosencephaly/ethnology , Humans , Male , Molecular Typing , Phenotype , Racial Groups , Sequence Analysis, DNA
14.
Eur J Med Genet ; 55(10): 510-4, 2012 Oct.
Article En | MEDLINE | ID: mdl-22750566

We have applied a GWAS to 40 consanguineous families segregating cases of non-syndromic cleft lip with or without cleft palate (NS CL/P) (a total of 160 affected and unaffected individuals) in order to trace potential recessive loci that confer susceptibility to this common facial malformation. Pedigree-based association test (PBAT) analyses reported nominal evidence of association and linkage over SNP markers located at 11q25 (rs4937877, P = 2.7 × 10(-6)), 19p12 (rs4324267, P = 1.6 × 10(-5)), 5q14.1 (rs4588572, P-value = 3.36 × 10(-5)), and 15q21.1 (rs4774497, P = 1.08 × 10(-4)). Using the Versatile Gene-Based Association Study to complement the PBAT results, we found clusters of markers located at chromosomes 19p12, 11q25, and 8p23.2 overcome the threshold for GWAS significance (P < 1 × 10(-7)). From this study, new recessive loci implicated in NS CL/P include: B3GAT1, GLB1L2, ZNF431, ZNF714, and CSMD1, even though the functional association with the genesis of NS CL/P remains to be elucidated. These results emphasize the importance of using homogeneous populations, phenotypes, and family structures for GWAS combined with gene-based association analyses, and should encourage. other researchers to evaluate these genes on independent patient samples affected by NS CL/P.


Cleft Lip/genetics , Cleft Palate/genetics , Genes, Recessive , Brain/abnormalities , Case-Control Studies , Cleft Lip/epidemiology , Cleft Palate/epidemiology , DNA-Binding Proteins/genetics , Female , Genetic Markers , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Glucuronosyltransferase/genetics , Glycoside Hydrolases/genetics , Humans , Male , Membrane Proteins/genetics , Pedigree , Transcription Factors/genetics , Tumor Suppressor Proteins
15.
Mol Genet Metab ; 105(3): 382-9, 2012 Mar.
Article En | MEDLINE | ID: mdl-22264778

Whole genome sequence data for small pedigrees has been shown to provide sufficient information to resolve detailed haplotypes in small pedigrees. Using such information, recombinations can be mapped onto chromosomes, compared with the segregation of a disease of interest and used to filter genome sequence variants. We now show that relatively inexpensive SNP array data from small pedigrees can be used in a similar manner to provide a means of identifying regions of interest in exome sequencing projects. We demonstrate that in those situations where one can assume complete penetrance and parental DNA is available, SNP recombination mapping using Boolean logic identifies chromosomal regions identical to those detected by multipoint linkage using microsatellites but with much less computation. We further show that this approach is successful because the probability of a double crossover between informative SNP loci is negligible. Our observations provide a rationale for using SNP arrays and recombination mapping as a rapid and cost-effective means of incorporating chromosome segregation information into exome sequencing projects intended for disease-gene identification.


Chromosome Mapping/methods , Exome/genetics , Polymorphism, Single Nucleotide , Recombination, Genetic , Base Sequence , DNA/analysis , Female , Genotype , Humans , Male , Oligonucleotide Array Sequence Analysis , Pedigree , Sequence Analysis, DNA
16.
Int J Cancer ; 129(10): 2400-7, 2011 Nov 15.
Article En | MEDLINE | ID: mdl-21207418

Genome-wide linkage studies have been used to localize rare and highly penetrant prostate cancer (PRCA) susceptibility genes. Linkage studies performed in different ethnic backgrounds and populations have been somewhat disparate, resulting in multiple, often irreproducible signals because of genetic heterogeneity and high sporadic background of the disease. Our first genome-wide linkage study and subsequent fine-mapping study of Finnish hereditary prostate cancer (HPC) families gave evidence of linkage to one region. Here, we conducted subsequent scans with microsatellites and SNPs in a total of 69 Finnish HPC families. GENEHUNTER-PLUS was used for parametric and nonparametric analyses. Our microsatellite genome-wide linkage study provided evidence of linkage to 17q12-q23, with a heterogeneity LOD (HLOD) score of 3.14 in a total of 54 of the 69 families. Genome-wide SNP analysis of 59 of the 69 families gave a highest HLOD score of 3.40 at 2q37.3 under a dominant high penetrance model. Analyzing all 69 families by combining microsatellite and SNP maps also yielded HLOD scores of > 3.3 in two regions (2q37.3 and 17q12-q21.3). These significant linkage peaks on chromosome 2 and 17 confirm previous linkage evidence of a locus on 17q from other populations and provide a basis for continued research into genetic factors involved in PRCA. Fine-mapping analysis of these regions is ongoing and candidate genes at linked loci are currently under analysis.


Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 2 , Genetic Linkage , Prostatic Neoplasms/genetics , Chromosome Mapping , Finland , Genetic Predisposition to Disease , Humans , Male , Microsatellite Repeats
17.
Blood ; 116(23): 4990-5001, 2010 Dec 02.
Article En | MEDLINE | ID: mdl-20709904

Gray platelet syndrome (GPS) is an inherited bleeding disorder characterized by macrothrombocytopenia and absence of platelet α-granules resulting in typical gray platelets on peripheral smears. GPS is associated with a bleeding tendency, myelofibrosis, and splenomegaly. Reports on GPS are limited to case presentations. The causative gene and underlying pathophysiology are largely unknown. We present the results of molecular genetic analysis of 116 individuals including 25 GPS patients from 14 independent families as well as novel clinical data on the natural history of the disease. The mode of inheritance was autosomal recessive (AR) in 11 and indeterminate in 3 families. Using genome-wide linkage analysis, we mapped the AR-GPS gene to a 9.4-Mb interval on 3p21.1-3p22.1, containing 197 protein-coding genes. Sequencing of 1423 (69%) of the 2075 exons in the interval did not identify the GPS gene. Long-term follow-up data demonstrated the progressive nature of the thrombocytopenia and myelofibrosis of GPS resulting in fatal hemorrhages in some patients. We identified high serum vitamin B(12) as a consistent, novel finding in GPS. Chromosome 3p21.1-3p22.1 has not been previously linked to a platelet disorder; identification of the GPS gene will likely lead to the discovery of novel components of platelet organelle biogenesis. This study is registered at www.clinicaltrials.gov as NCT00069680 and NCT00369421.


Chromosomes, Human, Pair 3/genetics , Gray Platelet Syndrome/genetics , Gray Platelet Syndrome/physiopathology , Adolescent , Adult , Blood Platelets/ultrastructure , Cell Separation , Child , Child, Preschool , DNA Mutational Analysis , Female , Flow Cytometry , Genetic Linkage , Genome-Wide Association Study , Gray Platelet Syndrome/blood , Humans , Male , Microsatellite Repeats , Microscopy, Electron, Transmission , Middle Aged , Neutrophils/ultrastructure , Pedigree , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Vitamin B 12/blood , Young Adult
18.
PLoS Genet ; 6(3): e1000870, 2010 Mar 05.
Article En | MEDLINE | ID: mdl-20221250

Papillorenal syndrome (PRS, also known as renal-coloboma syndrome) is an autosomal dominant disease characterized by potentially-blinding congenital optic nerve excavation and congenital kidney abnormalities. Many patients with PRS have mutations in the paired box transcription factor gene, PAX2. Although most mutations in PAX2 are predicted to result in complete loss of one allele's function, three missense mutations have been reported, raising the possibility that more subtle alterations in PAX2 function may be disease-causing. To date, the molecular behaviors of these mutations have not been explored. We describe a novel mouse model of PRS due to a missense mutation in a highly-conserved threonine residue in the paired domain of Pax2 (p.T74A) that recapitulates the ocular and kidney findings of patients. This mutation is in the Pax2 paired domain at the same location as two human missense mutations. We show that all three missense mutations disrupt potentially critical hydrogen bonds in atomic models and result in reduced Pax2 transactivation, but do not affect nuclear localization, steady state mRNA levels, or the ability of Pax2 to bind its DNA consensus sequence. Moreover, these mutations show reduced steady-state levels of Pax2 protein in vitro and (for p.T74A) in vivo, likely by reducing protein stability. These results suggest that hypomorphic alleles of PAX2/Pax2 can lead to significant disease in humans and mice.


Abnormalities, Multiple/genetics , Alleles , Mutation, Missense/genetics , PAX2 Transcription Factor/genetics , Amino Acid Sequence , Animals , Cell Line , Cerebellum/pathology , DNA/metabolism , Embryo, Mammalian/pathology , Eye/pathology , Gene Expression Regulation, Developmental , Humans , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , PAX2 Transcription Factor/chemistry , PAX2 Transcription Factor/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Structural Homology, Protein , Syndrome , Time Factors
19.
Am J Med Genet A ; 149A(7): 1382-91, 2009 Jul.
Article En | MEDLINE | ID: mdl-19530184

Smith-Magenis syndrome (SMS) is a disorder characterized by multiple congenital anomalies and behavior problems, including abnormal sleep patterns. It is most commonly due to a 3.5 Mb interstitial deletion of chromosome 17 band p11.2. Secretion of melatonin, a hormone produced by the pineal gland, is the body's signal for nighttime darkness. Published reports of 24-hr melatonin secretion patterns in two independent SMS cohorts (US and France) document an inverted endogenous melatonin pattern in virtually all cases (96%), suggesting that this finding is pathognomic for the syndrome. We report on a woman with SMS due to an atypical large proximal deletion ( approximately 6Mb; cen<->TNFRSFproteinB) of chromosome band (17)(p11.2p11.2) who presents with typical sleep disturbances but a normal pattern of melatonin secretion. We further describe a melatonin light suppression test in this patient. This is the second reported patient with a normal endogenous melatonin rhythm in SMS associated with an atypical large deletion. These two patients are significant because they suggest that the sleep disturbances in SMS cannot be solely attributed to the abnormal diurnal melatonin secretion versus the normal nocturnal pattern.


Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, Pair 17 , Melatonin/metabolism , Sleep Initiation and Maintenance Disorders/genetics , Abnormalities, Multiple/metabolism , Female , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Medical History Taking , Retrospective Studies , Sleep Initiation and Maintenance Disorders/complications , Sleep Initiation and Maintenance Disorders/diagnosis , Syndrome , Young Adult
20.
N Engl J Med ; 359(9): 918-27, 2008 Aug 28.
Article En | MEDLINE | ID: mdl-18753648

BACKGROUND: Brain-derived neurotrophic factor (BDNF) has been found to be important in energy homeostasis in animal models, but little is known about its role in energy balance in humans. Heterozygous, variably sized, contiguous gene deletions causing haploinsufficiency of the WT1 and PAX6 genes on chromosome 11p13, approximately 4 Mb centromeric to BDNF (11p14.1), result in the Wilms' tumor, aniridia, genitourinary anomalies, and mental retardation (WAGR) syndrome. Hyperphagia and obesity were observed in a subgroup of patients with the WAGR syndrome. We hypothesized that the subphenotype of obesity in the WAGR syndrome is attributable to deletions that induce haploinsufficiency of BDNF. METHODS: We studied the relationship between genotype and body-mass index (BMI) in 33 patients with the WAGR syndrome who were recruited through the International WAGR Syndrome Association. The extent of each deletion was determined with the use of oligonucleotide comparative genomic hybridization. RESULTS: Deletions of chromosome 11p in the patients studied ranged from 1.0 to 26.5 Mb; 58% of the patients had heterozygous BDNF deletions. These patients had significantly higher BMI z scores throughout childhood than did patients with intact BDNF (mean [+/-SD] z score at 8 to 10 years of age, 2.08+/-0.45 in patients with heterozygous BDNF deletions vs. 0.88+/-1.28 in patients without BDNF deletions; P=0.03). By 10 years of age, 100% of the patients with heterozygous BDNF deletions (95% confidence interval [CI], 77 to 100) were obese (BMI > or = 95th percentile for age and sex) as compared with 20% of persons without BDNF deletions (95% CI, 3 to 56; P<0.001). The critical region for childhood-onset obesity in the WAGR syndrome was located within 80 kb of exon 1 of BDNF. Serum BDNF concentrations were approximately 50% lower among the patients with heterozygous BDNF deletions (P=0.001). CONCLUSIONS: Among persons with the WAGR syndrome, BDNF haploinsufficiency is associated with lower levels of serum BDNF and with childhood-onset obesity; thus, BDNF may be important for energy homeostasis in humans.


Brain-Derived Neurotrophic Factor/genetics , Gene Deletion , Obesity/genetics , WAGR Syndrome/genetics , Adolescent , Adult , Body Mass Index , Brain-Derived Neurotrophic Factor/blood , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 11 , Energy Metabolism/genetics , Female , Genotype , Haplotypes , Homeostasis/genetics , Humans , Hyperphagia/etiology , Male , Microsatellite Repeats , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Pain Measurement , Surveys and Questionnaires , WAGR Syndrome/blood , WAGR Syndrome/complications
...