Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Cell ; 174(6): 1406-1423.e16, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193113

ABSTRACT

Probiotics are widely prescribed for prevention of antibiotics-associated dysbiosis and related adverse effects. However, probiotic impact on post-antibiotic reconstitution of the gut mucosal host-microbiome niche remains elusive. We invasively examined the effects of multi-strain probiotics or autologous fecal microbiome transplantation (aFMT) on post-antibiotic reconstitution of the murine and human mucosal microbiome niche. Contrary to homeostasis, antibiotic perturbation enhanced probiotics colonization in the human mucosa but only mildly improved colonization in mice. Compared to spontaneous post-antibiotic recovery, probiotics induced a markedly delayed and persistently incomplete indigenous stool/mucosal microbiome reconstitution and host transcriptome recovery toward homeostatic configuration, while aFMT induced a rapid and near-complete recovery within days of administration. In vitro, Lactobacillus-secreted soluble factors contributed to probiotics-induced microbiome inhibition. Collectively, potential post-antibiotic probiotic benefits may be offset by a compromised gut mucosal recovery, highlighting a need of developing aFMT or personalized probiotic approaches achieving mucosal protection without compromising microbiome recolonization in the antibiotics-perturbed host.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Probiotics/administration & dosage , Adolescent , Adult , Aged , Animals , Fecal Microbiota Transplantation , Feces/microbiology , Female , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Lactobacillus/drug effects , Lactobacillus/genetics , Lactobacillus/isolation & purification , Lactococcus/genetics , Lactococcus/isolation & purification , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA, Ribosomal, 16S/analysis , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Young Adult
2.
Cell ; 167(6): 1495-1510.e12, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912059

ABSTRACT

The intestinal microbiota undergoes diurnal compositional and functional oscillations that affect metabolic homeostasis, but the mechanisms by which the rhythmic microbiota influences host circadian activity remain elusive. Using integrated multi-omics and imaging approaches, we demonstrate that the gut microbiota features oscillating biogeographical localization and metabolome patterns that determine the rhythmic exposure of the intestinal epithelium to different bacterial species and their metabolites over the course of a day. This diurnal microbial behavior drives, in turn, the global programming of the host circadian transcriptional, epigenetic, and metabolite oscillations. Surprisingly, disruption of homeostatic microbiome rhythmicity not only abrogates normal chromatin and transcriptional oscillations of the host, but also incites genome-wide de novo oscillations in both intestine and liver, thereby impacting diurnal fluctuations of host physiology and disease susceptibility. As such, the rhythmic biogeography and metabolome of the intestinal microbiota regulates the temporal organization and functional outcome of host transcriptional and epigenetic programs.


Subject(s)
Circadian Rhythm , Colon/microbiology , Gastrointestinal Microbiome , Transcriptome , Animals , Chromatin/metabolism , Colon/metabolism , Germ-Free Life , Liver/metabolism , Mice , Microscopy, Electron, Scanning
3.
Cell ; 163(6): 1428-43, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26638072

ABSTRACT

Host-microbiome co-evolution drives homeostasis and disease susceptibility, yet regulatory principles governing the integrated intestinal host-commensal microenvironment remain obscure. While inflammasome signaling participates in these interactions, its activators and microbiome-modulating mechanisms are unknown. Here, we demonstrate that the microbiota-associated metabolites taurine, histamine, and spermine shape the host-microbiome interface by co-modulating NLRP6 inflammasome signaling, epithelial IL-18 secretion, and downstream anti-microbial peptide (AMP) profiles. Distortion of this balanced AMP landscape by inflammasome deficiency drives dysbiosis development. Upon fecal transfer, colitis-inducing microbiota hijacks this microenvironment-orchestrating machinery through metabolite-mediated inflammasome suppression, leading to distorted AMP balance favoring its preferential colonization. Restoration of the metabolite-inflammasome-AMP axis reinstates a normal microbiota and ameliorates colitis. Together, we identify microbial modulators of the NLRP6 inflammasome and highlight mechanisms by which microbiome-host interactions cooperatively drive microbial community stability through metabolite-mediated innate immune modulation. Therefore, targeted "postbiotic" metabolomic intervention may restore a normal microenvironment as treatment or prevention of dysbiosis-driven diseases.


Subject(s)
Colon/immunology , Colon/microbiology , Inflammasomes/immunology , Microbiota , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Antimicrobial Cationic Peptides , Colitis/chemically induced , Colitis/drug therapy , Colon/metabolism , Dysbiosis/metabolism , Germ-Free Life , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Interleukin-18/immunology , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/genetics , Taurine/administration & dosage
4.
Cell ; 163(5): 1079-1094, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26590418

ABSTRACT

Elevated postprandial blood glucose levels constitute a global epidemic and a major risk factor for prediabetes and type II diabetes, but existing dietary methods for controlling them have limited efficacy. Here, we continuously monitored week-long glucose levels in an 800-person cohort, measured responses to 46,898 meals, and found high variability in the response to identical meals, suggesting that universal dietary recommendations may have limited utility. We devised a machine-learning algorithm that integrates blood parameters, dietary habits, anthropometrics, physical activity, and gut microbiota measured in this cohort and showed that it accurately predicts personalized postprandial glycemic response to real-life meals. We validated these predictions in an independent 100-person cohort. Finally, a blinded randomized controlled dietary intervention based on this algorithm resulted in significantly lower postprandial responses and consistent alterations to gut microbiota configuration. Together, our results suggest that personalized diets may successfully modify elevated postprandial blood glucose and its metabolic consequences. VIDEO ABSTRACT.


Subject(s)
Algorithms , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Postprandial Period , Diabetes Mellitus, Type 2/diet therapy , Diabetes Mellitus, Type 2/microbiology , Diet, Diabetic , Gastrointestinal Microbiome , Humans , Smartphone
5.
Cell ; 159(3): 514-29, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417104

ABSTRACT

All domains of life feature diverse molecular clock machineries that synchronize physiological processes to diurnal environmental fluctuations. However, no mechanisms are known to cross-regulate prokaryotic and eukaryotic circadian rhythms in multikingdom ecosystems. Here, we show that the intestinal microbiota, in both mice and humans, exhibits diurnal oscillations that are influenced by feeding rhythms, leading to time-specific compositional and functional profiles over the course of a day. Ablation of host molecular clock components or induction of jet lag leads to aberrant microbiota diurnal fluctuations and dysbiosis, driven by impaired feeding rhythmicity. Consequently, jet-lag-induced dysbiosis in both mice and humans promotes glucose intolerance and obesity that are transferrable to germ-free mice upon fecal transplantation. Together, these findings provide evidence of coordinated metaorganism diurnal rhythmicity and offer a microbiome-dependent mechanism for common metabolic disturbances in humans with aberrant circadian rhythms, such as those documented in shift workers and frequent flyers.


Subject(s)
Circadian Clocks , Circadian Rhythm , Glucose Intolerance , Microbiota , Animals , Dysbiosis/microbiology , Dysbiosis/physiopathology , Feeding Behavior , Homeostasis , Humans , Jet Lag Syndrome/physiopathology , Metabolic Diseases/microbiology , Metabolic Diseases/physiopathology , Mice , Obesity/metabolism , Sleep
6.
Nature ; 588(7836): 135-140, 2020 12.
Article in English | MEDLINE | ID: mdl-33177712

ABSTRACT

The serum metabolome contains a plethora of biomarkers and causative agents of various diseases, some of which are endogenously produced and some that have been taken up from the environment1. The origins of specific compounds are known, including metabolites that are highly heritable2,3, or those that are influenced by the gut microbiome4, by lifestyle choices such as smoking5, or by diet6. However, the key determinants of most metabolites are still poorly understood. Here we measured the levels of 1,251 metabolites in serum samples from a unique and deeply phenotyped healthy human cohort of 491 individuals. We applied machine-learning algorithms to predict metabolite levels in held-out individuals on the basis of host genetics, gut microbiome, clinical parameters, diet, lifestyle and anthropometric measurements, and obtained statistically significant predictions for more than 76% of the profiled metabolites. Diet and microbiome had the strongest predictive power, and each explained hundreds of metabolites-in some cases, explaining more than 50% of the observed variance. We further validated microbiome-related predictions by showing a high replication rate in two geographically independent cohorts7,8 that were not available to us when we trained the algorithms. We used feature attribution analysis9 to reveal specific dietary and bacterial interactions. We further demonstrate that some of these interactions might be causal, as some metabolites that we predicted to be positively associated with bread were found to increase after a randomized clinical trial of bread intervention. Overall, our results reveal potential determinants of more than 800 metabolites, paving the way towards a mechanistic understanding of alterations in metabolites under different conditions and to designing interventions for manipulating the levels of circulating metabolites.


Subject(s)
Diet , Gastrointestinal Microbiome/physiology , Metabolome/genetics , Serum/metabolism , Adult , Bread , Cohort Studies , Female , Healthy Volunteers , Humans , Life Style , Machine Learning , Male , Metabolomics , Middle Aged , Non-alcoholic Fatty Liver Disease/genetics , Oxygenases/genetics , Reference Standards , Reproducibility of Results , Seasons
7.
Am J Hum Genet ; 109(6): 981-988, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35659933

ABSTRACT

The underrepresentation of non-European ancestry groups in current genomic databases complicates interpretation of their genetic test results, yielding a much higher prevalence of variants of uncertain significance (VUSs). Such VUS findings can frustrate the goals of genetic testing, create anxiety in patients, and lead to unnecessary medical interventions. Approaches to addressing underrepresentation of people with genetic ancestries other than European are being undertaken by broad-based recruitment efforts. However, some underrepresented groups have concerns that might preclude participation in such efforts. We describe here two initiatives aimed at meeting the needs of underrepresented ancestry groups in genomic datasets. The two communities, the Sephardi Jewish community in New York and First Peoples of Canada, have very different concerns about contributing to genomic research and datasets. Sephardi concerns focus on the possible negative effects of genetic findings on the marriage prospects of family members. Canadian Indigenous populations seek control over the research uses to which their genetic data would be put. Both cases involve targeted efforts to respond to the groups' concerns; these efforts include governance models aimed at ensuring that the data are used primarily to inform clinical test analyses and at achieving successful engagement and participation of community members. We suggest that these initiatives could provide models for other ancestral groups seeking to improve the accuracy and utility of clinical genetic testing while respecting the underlying preferences and values of community members with regard to the use of their genetic data.


Subject(s)
Ethnicity , Genetic Testing , Canada , Ethnicity/genetics , Family , Genomics , Humans
8.
Nature ; 568(7750): 43-48, 2019 04.
Article in English | MEDLINE | ID: mdl-30918406

ABSTRACT

Differences in the presence of even a few genes between otherwise identical bacterial strains may result in critical phenotypic differences. Here we systematically identify microbial genomic structural variants (SVs) and find them to be prevalent in the human gut microbiome across phyla and to replicate in different cohorts. SVs are enriched for CRISPR-associated and antibiotic-producing functions and depleted from housekeeping genes, suggesting that they have a role in microbial adaptation. We find multiple associations between SVs and host disease risk factors, many of which replicate in an independent cohort. Exploring genes that are clustered in the same SV, we uncover several possible mechanistic links between the microbiome and its host, including a region in Anaerostipes hadrus that encodes a composite inositol catabolism-butyrate biosynthesis pathway, the presence of which is associated with lower host metabolic disease risk. Overall, our results uncover a nascent layer of variability in the microbiome that is associated with microbial adaptation and host health.


Subject(s)
Bacteria/genetics , Disease Susceptibility/microbiology , Gastrointestinal Microbiome/genetics , Genes, Bacterial/genetics , Genetic Variation , Health , Host Microbial Interactions/genetics , Adaptation, Physiological/genetics , Bacteria/classification , Bacteria/growth & development , Bacteria/metabolism , Butyrates/metabolism , Cohort Studies , Ecosystem , Eubacterium/genetics , Eubacterium/metabolism , Feces/microbiology , Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Humans , Inositol/metabolism , Metagenomics , Microbial Viability/genetics , Risk Factors
9.
Nature ; 555(7695): 210-215, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29489753

ABSTRACT

Human gut microbiome composition is shaped by multiple factors but the relative contribution of host genetics remains elusive. Here we examine genotype and microbiome data from 1,046 healthy individuals with several distinct ancestral origins who share a relatively common environment, and demonstrate that the gut microbiome is not significantly associated with genetic ancestry, and that host genetics have a minor role in determining microbiome composition. We show that, by contrast, there are significant similarities in the compositions of the microbiomes of genetically unrelated individuals who share a household, and that over 20% of the inter-person microbiome variability is associated with factors related to diet, drugs and anthropometric measurements. We further demonstrate that microbiome data significantly improve the prediction accuracy for many human traits, such as glucose and obesity measures, compared to models that use only host genetic and environmental data. These results suggest that microbiome alterations aimed at improving clinical outcomes may be carried out across diverse genetic backgrounds.


Subject(s)
Diet/statistics & numerical data , Environment , Family Characteristics , Gastrointestinal Microbiome/genetics , Life Style , Adolescent , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Gene-Environment Interaction , Glucose/metabolism , Healthy Volunteers , Heredity/genetics , Humans , Israel , Male , Middle Aged , Obesity/metabolism , Phenotype , Polymorphism, Single Nucleotide/genetics , RNA, Bacterial/analysis , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/analysis , Reproducibility of Results , Twin Studies as Topic , Twins/genetics , Young Adult
10.
Nucleic Acids Res ; 50(3): 1661-1672, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35048966

ABSTRACT

CRISPR-Cas systems provide prokaryotic organisms with an adaptive defense mechanism that acquires immunological memories of infections. This is accomplished by integration of short fragments from the genome of invaders such as phages and plasmids, called 'spacers', into the CRISPR locus of the host. Depending on their genetic composition, CRISPR-Cas systems can be classified into six types, I-VI, however spacer acquisition has been extensively studied only in type I and II systems. Here, we used an inducible spacer acquisition assay to study this process in the type III-A CRISPR-Cas system of Staphylococcus epidermidis, in the absence of phage selection. Similarly to type I and II spacer acquisition, this type III system uses Cas1 and Cas2 to preferentially integrate spacers from the chromosomal terminus and free dsDNA ends produced after DNA breaks, in a manner that is enhanced by the AddAB DNA repair complex. Surprisingly, a different mode of spacer acquisition from rRNA and tRNA loci, which spans only the transcribed sequences of these genes and is not enhanced by AddAB, was also detected. Therefore, our findings reveal both common mechanistic principles that may be conserved in all CRISPR-Cas systems, as well as unique and intriguing features of type III spacer acquisition.


Subject(s)
Staphylococcus epidermidis/genetics , Bacteriophages/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Plasmids/genetics , Staphylococcus epidermidis/metabolism , Staphylococcus epidermidis/virology
11.
Am J Hum Genet ; 106(5): 623-631, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32275884

ABSTRACT

Nucleoporins (NUPs) are an essential component of the nuclear-pore complex, which regulates nucleocytoplasmic transport of macromolecules. Pathogenic variants in NUP genes have been linked to several inherited human diseases, including a number with progressive neurological degeneration. We present six affected individuals with bi-allelic truncating variants in NUP188 and strikingly similar phenotypes and clinical courses, representing a recognizable genetic syndrome; the individuals are from four unrelated families. Key clinical features include congenital cataracts, hypotonia, prenatal-onset ventriculomegaly, white-matter abnormalities, hypoplastic corpus callosum, congenital heart defects, and central hypoventilation. Characteristic dysmorphic features include small palpebral fissures, a wide nasal bridge and nose, micrognathia, and digital anomalies. All affected individuals died as a result of respiratory failure, and five of them died within the first year of life. Nuclear import of proteins was decreased in affected individuals' fibroblasts, supporting a possible disease mechanism. CRISPR-mediated knockout of NUP188 in Drosophila revealed motor deficits and seizure susceptibility, partially recapitulating the neurological phenotype seen in affected individuals. Removal of NUP188 also resulted in aberrant dendrite tiling, suggesting a potential role of NUP188 in dendritic development. Two of the NUP188 pathogenic variants are enriched in the Ashkenazi Jewish population in gnomAD, a finding we confirmed with a separate targeted population screen of an international sampling of 3,225 healthy Ashkenazi Jewish individuals. Taken together, our results implicate bi-allelic loss-of-function NUP188 variants in a recessive syndrome characterized by a distinct neurologic, ophthalmologic, and facial phenotype.


Subject(s)
Alleles , Brain/abnormalities , Drosophila Proteins/genetics , Eye Abnormalities/genetics , Heart Defects, Congenital/genetics , Loss of Function Mutation/genetics , Nuclear Pore Complex Proteins/genetics , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Child, Preschool , Dendrites/metabolism , Dendrites/pathology , Drosophila melanogaster , Eye Abnormalities/mortality , Female , Fibroblasts , Genes, Recessive , Heart Defects, Congenital/mortality , Humans , Infant , Infant, Newborn , Jews/genetics , Male , Nuclear Pore Complex Proteins/deficiency , Seizures/metabolism , Syndrome , beta Karyopherins/metabolism
12.
Mol Genet Genomics ; 297(4): 925-933, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35488049

ABSTRACT

Complex chromosomal rearrangements (CCRs), a class of structural variants (SVs) involving more than two chromosome breaks, were classically thought to be extremely rare. As advanced technologies become more available, it has become apparent that CCRs are more common than formerly thought, and are a substantial cause of genetic disorders. We attempted a novel approach for solving the mechanism of challenging CCRs, which involve repetitive sequences, by precisely identifying sequence-level changes and their order. Chromosomal microarray (CMA) and FISH analyses were used for interpretation of SVs detected by whole exome sequencing (WES). Breakpoint junctions were analyzed by Nanopore sequencing, a novel long-read whole genome sequencing tool. A large deletion identified by WES, encompassing the FOXF1 enhancer, was the cause of alveolar capillary dysplasia and respiratory insufficiency, resulting in perinatal death. CMA analysis of the newborn's mother revealed two duplications encompassing the deleted region in the proband, raising our hypothesis that the deletion resulted from the mother's CCR. Breakpoint junctions of complex SVs were determined at the nucleotide level using Nanopore long-read sequencing. According to sequencing results of breakpoint junctions, the CCR in the newborn was considered the consequence of at least one double-strand break during meiosis, and reassembly of DNA fragments by intra-chromosomal homologous recombination. Our comprehensive approach, combining cytogenetics and long-read sequencing, enabled delineation of the exact breakpoints in a challenging CCR, and proposal of a mechanism in which it arises. We suggest applying our integrative approach combining technologies for deciphering future challenging CCRs, enabling risk assessment in families.


Subject(s)
Chromosome Aberrations , Genome , Chromosomes , Cytogenetic Analysis , Female , Genomics , Humans , Pregnancy
13.
Am J Med Genet A ; 188(5): 1420-1425, 2022 05.
Article in English | MEDLINE | ID: mdl-35075769

ABSTRACT

Variants involving TBX4 are associated with a wide variety of disorders, including pulmonary arterial hypertension, ischiocoxopodopatellar syndrome (ICPPS)/small patella syndrome (SPS), lethal lung developmental disorders (LLDDs) in neonates, heart defects, and prenatally lethal posterior amelia with pelvic and pulmonary hypoplasia syndrome. The objective of our study was to elucidate the wide variable phenotypic expressivity and incomplete penetrance in a three-generation family with a truncating variant in TBX4. In addition to exome and genome sequencing analyses, a candidate noncoding regulatory single nucleotide variant (SNV) within the lung-specific TBX4 enhancer was functionally tested using an in vitro luciferase reporter assay. A heterozygous frameshift variant c.1112dup (p.Pro372Serfs*14) in TBX4 was identified in patients with mild interstitial lung disease (1), bronchiolitis obliterans (1), recurrent pneumothorax (1), ICPPS/SPS (1), LLDD (2), and in unaffected individuals (4). In two deceased neonates with LLDD, we identified a noncoding SNV rs62069651-C located in trans to the mutated TBX4 allele that reduced the TBX4 promoter activity by 63% in the reporter assay. Our findings provide a functional evidence for the recently reported model of complex compound inheritance in which both TBX4 coding and in trans noncoding hypomorphic variants in the lung-specific enhancer of TBX4 contribute to LLDD.


Subject(s)
Lung Diseases , Respiratory System Abnormalities , Bone Diseases, Developmental , Hip/abnormalities , Humans , Infant, Newborn , Ischium/abnormalities , Lung/abnormalities , Lung Diseases/genetics , Patella/abnormalities , T-Box Domain Proteins/genetics
14.
Genet Med ; 23(7): 1334-1340, 2021 07.
Article in English | MEDLINE | ID: mdl-33772222

ABSTRACT

PURPOSE: We previously developed Haploseek, a method for comprehensive preimplantation genetic testing (PGT). However, some key features were missing, and the method has not yet been systematically validated. METHODS: We extended Haploseek to incorporate DNA from embryo grandparents and to allow testing of variants on chromosome X or in regions where parents share common haplotypes. We then validated Haploseek on 151 embryo biopsies from 27 clinical PGT cases. We sequenced all biopsies to low coverage (0.2×), and performed single-nucleotide polymorphism (SNP) microarray genotyping on the embryos' parents and siblings/grandparents. We used the extended Haploseek to predict chromosome copy-number variants (CNVs) and relevant variant-flanking haplotypes in each embryo. We validated haplotype predictions for each clinical sample against polymerase chain reaction (PCR)-based PGT case results, and CNV predictions against established commercial kits. RESULTS: For each of the 151 embryo biopsies, all Haploseek-derived haplotypes and CNVs were concordant with clinical PGT results. The cases included 17 autosomal dominant, 5 autosomal recessive, and 3 X-linked monogenic disorders. In addition, we evaluated 1 Robertsonian and 2 reciprocal translocations, and 17 cases of chromosome copy-number counting were performed. CONCLUSION: Our results demonstrate that Haploseek is clinically accurate and fit for all standard clinical PGT applications.


Subject(s)
Preimplantation Diagnosis , DNA Copy Number Variations/genetics , Female , Genetic Testing , Haplotypes , Humans , Pregnancy , Translocation, Genetic
15.
Nature ; 514(7521): 181-6, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25231862

ABSTRACT

Non-caloric artificial sweeteners (NAS) are among the most widely used food additives worldwide, regularly consumed by lean and obese individuals alike. NAS consumption is considered safe and beneficial owing to their low caloric content, yet supporting scientific data remain sparse and controversial. Here we demonstrate that consumption of commonly used NAS formulations drives the development of glucose intolerance through induction of compositional and functional alterations to the intestinal microbiota. These NAS-mediated deleterious metabolic effects are abrogated by antibiotic treatment, and are fully transferrable to germ-free mice upon faecal transplantation of microbiota configurations from NAS-consuming mice, or of microbiota anaerobically incubated in the presence of NAS. We identify NAS-altered microbial metabolic pathways that are linked to host susceptibility to metabolic disease, and demonstrate similar NAS-induced dysbiosis and glucose intolerance in healthy human subjects. Collectively, our results link NAS consumption, dysbiosis and metabolic abnormalities, thereby calling for a reassessment of massive NAS usage.


Subject(s)
Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Glucose Intolerance/chemically induced , Glucose Intolerance/microbiology , Microbiota/drug effects , Sweetening Agents/adverse effects , Animals , Anti-Bacterial Agents/pharmacology , Aspartame/adverse effects , Body Weight/drug effects , Diet, High-Fat , Dietary Fats/pharmacology , Feces/microbiology , Female , Germ-Free Life , Glucose/metabolism , Glucose Intolerance/metabolism , Humans , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/metabolism , Metabolic Syndrome/microbiology , Mice , Mice, Inbred C57BL , Saccharin/administration & dosage , Saccharin/adverse effects , Sucrose/adverse effects , Sucrose/analogs & derivatives , Waist-Hip Ratio
16.
Eur J Pediatr ; 179(12): 1843-1849, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32524198

ABSTRACT

Our aim was to evaluate the performance of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS), routinely used in the microbiology laboratory for bacterial identification, for bacterial typing in the setting of extended spectrum beta-lactamase producing Klebsiella pneumoniae (ESBL-KP) outbreak in the neonatal intensive care unit (NICU). Isolates from a 2011 outbreak in the NICU were retrieved from frozen stocks and analyzed by MALDI-TOF. The MALDI typing was compared with core genome multilocus sequence typing (cg-MLST). MALDI typing divided the 33 outbreak isolates into 2 clones: sequence type (ST)-290 and 405. These results were in complete agreement with cg-MLST results. The differentiation of the outbreak isolates into two clones correlated with the patients' location in the NICU, but also with their place of residence.Conclusion: Here, we show that MALDI-TOF MS, which has been integrated into the microbiology laboratory workflow for microbial species identification, can be secondarily used for epidemiological typing at no added cost. What is Known: • Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) is now routinely used in the microbiology laboratory for bacterial identification What is New: • MALDI typing was used for outbreak investigation in the NICU and divided the outbreak isolates into two clones • MALDI-TOF MS may be secondarily used for epidemiological typing at no added cost.


Subject(s)
Intensive Care Units, Neonatal , Klebsiella Infections , Klebsiella pneumoniae , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Disease Outbreaks , Humans , Infant, Newborn , Klebsiella Infections/diagnosis , Klebsiella pneumoniae/genetics , Multilocus Sequence Typing
17.
J Assist Reprod Genet ; 37(8): 1903-1912, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32462417

ABSTRACT

PURPOSE: The decision to undergo preimplantation genetic testing (PGT) entails a variety of personal and societal variables. Although PGT technology is widely accepted and used, few studies have queried the motives and concerns of PGT users; moreover, in-depth qualitative data regarding the PGT experience is scant. METHODS: In order to explore and analyze the experience, concerns, expectations, and attitudes toward the PGT technique and its implications, semi-structured interviews were conducted in a single tertiary medical center with 43 Israeli PGT users for HLA matching and autosomal dominant, autosomal recessive, and X-linked genetic disorders. RESULTS: The primary considerations in choosing PGT were prevention of birth of a child who would suffer a terminal or chronic disease as well as abrogation of a familial genetic condition. Religion played a decisive role in accepting PGT as an antenatal option. Regarding satisfaction with the PGT experience, many interviewees highlighted the need for greater attention to be given to potential stages of failure throughout the procedure and the need for emotional support. Our clinical results regarding implantation rate and cumulative live birth rate are 38-40% and 27-30%, respectively. CONCLUSION: This survey broadens understanding of the specialized needs of women, couples, and minority groups undergoing PGT and underscores the relevance of counseling services for PGT users.


Subject(s)
Embryo Implantation/genetics , Fertilization in Vitro , Genetic Testing/methods , Preimplantation Diagnosis , Adult , Aneuploidy , Birth Rate , Clinical Decision-Making , Embryo Implantation/physiology , Embryo Transfer/methods , Female , Humans , Mutation/genetics , Pregnancy
18.
Genet Med ; 21(6): 1390-1399, 2019 06.
Article in English | MEDLINE | ID: mdl-30449887

ABSTRACT

PURPOSE: To develop an economical, user-friendly, and accurate all-in-one next-generation sequencing (NGS)-based workflow for single-cell gene variant detection combined with comprehensive chromosome screening in a 24-hour workflow protocol. METHODS: We subjected single lymphoblast cells or blastomere/blastocyst biopsies from four different families to low coverage (0.3×-1.4×) genome sequencing. We combined copy-number variant (CNV) detection and whole-genome haplotype phase prediction via Haploseek, a novel, user-friendly analysis pipeline. We validated haplotype predictions for each sample by comparing with clinical preimplantation genetic diagnosis (PGD) case results or by single-nucleotide polymorphism (SNP) microarray analysis of bulk DNA from each respective lymphoblast culture donor. CNV predictions were validated by established commercial kits for single-cell CNV prediction. RESULTS: Haplotype phasing of the single lymphoblast/embryo biopsy sequencing data was highly concordant with relevant ground truth haplotypes in all samples/biopsies from all four families. In addition, whole-genome copy-number assessments were concordant with the results of a commercial kit. CONCLUSION: Our results demonstrate the establishment of a reliable method for all-in-one molecular and chromosomal diagnosis of single cells. Important features of the Haploseek pipeline include rapid sample processing, rapid sequencing, streamlined analysis, and user-friendly reporting, so as to expedite clinical PGD implementation.


Subject(s)
Genetic Testing/methods , Haplotypes/genetics , Preimplantation Diagnosis/methods , Aneuploidy , Biopsy , Blastocyst , Chromosomes , DNA Copy Number Variations/genetics , Female , Fertilization in Vitro , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Pregnancy
19.
Am J Med Genet A ; 179(10): 2144-2151, 2019 10.
Article in English | MEDLINE | ID: mdl-31287223

ABSTRACT

Warsaw breakage syndrome (WABS), caused by bi-allelic variants in the DDX11 gene, is a rare cohesinopathy characterized by pre- and postnatal growth retardation, microcephaly, intellectual disability, facial dysmorphia, and sensorineural hearing loss due to cochlear hypoplasia. The DDX11 gene codes for an iron-sulfur DNA helicase in the Superfamily 2 helicases and plays an important role in genomic stability and maintenance. Fourteen individuals with WABS have been previously reported in the medical literature. Affected individuals have been of various ethnic backgrounds with different pathogenic variants. We report two unrelated individuals of Ashkenazi Jewish descent affected with WABS, who are homozygous for the c.1763-1G>C variant in the DDX11 gene. Their phenotype is consistent with previously reported individuals. RNA studies showed that this variant causes an alternative splice acceptor site leading to a frameshift in the open reading frame. Carrier screening of the c.1763-1G>C variant in the Jewish population revealed a high carrier frequency of 1 in 68 in the Ashkenazi Jewish population. Due to the high carrier frequency and the low number of affected individuals, we hypothesize a high rate of miscarriage of homozygous fetuses and/or subfertility for carrier couples. If the carrier frequency is reproducible in additional Ashkenazi Jewish populations, we suggest including DDX11 to Ashkenazi Jewish carrier screening panels.


Subject(s)
Abnormalities, Multiple/genetics , Jews/genetics , Adolescent , Base Sequence , Child , Child, Preschool , Female , Genetic Testing , Heterozygote , Humans , Infant , Infant, Newborn , Male , Phenotype , RNA Splicing/genetics , Syndrome , Young Adult
20.
J Assist Reprod Genet ; 36(4): 727-739, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30617673

ABSTRACT

PURPOSE: Pre-implantation genetic diagnosis (PGD) for molecular disorders requires the construction of parental haplotypes. Classically, haplotype resolution ("phasing") is obtained by genotyping multiple polymorphic markers in both parents and at least one additional relative. However, this process is time-consuming, and immediate family members are not always available. The recent availability of massive genomic data for many populations promises to eliminate the needs for developing family-specific assays and for recruiting additional family members. In this study, we aimed to validate population-assisted haplotype phasing for PGD. METHODS: Targeted sequencing of CFTR gene variants and ~ 1700 flanking polymorphic SNPs (± 2 Mb) was performed on 54 individuals from 12 PGD families of (a) Full Ashkenazi (FA; n = 16), (b) mixed Ashkenazi (MA; n = 23 individuals with at least one Ashkenazi and one non-Ashkenazi grandparents), or (c) non-Ashkenazi (NA; n = 15) descent. Heterozygous genotype calls in each individual were phased using various whole genome reference panels and appropriate computational models. All computationally derived haplotype predictions were benchmarked against trio-based phasing. RESULTS: Using the Ashkenazi reference panel, phasing of FA was highly accurate (99.4% ± 0.2% accuracy); phasing of MA was less accurate (95.4% ± 4.5% accuracy); and phasing of NA was predictably low (83.4% ± 6.6% accuracy). Strikingly, for founder mutation carriers, our haplotyping approach facilitated near perfect phasing accuracy (99.9% ± 0.1% and 98.2% ± 2.8% accuracy for W1282X and delF508 carriers, respectively). CONCLUSIONS: Our results demonstrate the feasibility of replacing classical haplotype phasing with population-based phasing with uncompromised accuracy.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Genotype , Haplotypes/genetics , Preimplantation Diagnosis , Algorithms , Alleles , Female , Founder Effect , Heterozygote , Humans , Jews/genetics , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL