Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Meaghan K Russell is active.

Publication


Featured researches published by Meaghan K Russell.


Nature Genetics | 2007

Mutations in LRP2 , which encodes the multiligand receptor megalin, cause Donnai-Barrow and facio-oculo-acoustico-renal syndromes

Sibel Kantarci; Lihadh Al-Gazali; R. Sean Hill; Dian Donnai; Graeme C.M. Black; Eric Bieth; Nicolas Chassaing; Didier Lacombe; Koenraad Devriendt; Ahmad S. Teebi; Maria Loscertales; Caroline D. Robson; Tianming Liu; David T. MacLaughlin; Kristin M Noonan; Meaghan K Russell; Christopher A. Walsh; Patricia K. Donahoe; Barbara R. Pober

Donnai-Barrow syndrome is associated with agenesis of the corpus callosum, congenital diaphragmatic hernia, facial dysmorphology, ocular anomalies, sensorineural hearing loss and developmental delay. By studying multiplex families, we mapped this disorder to chromosome 2q23.3–31.1 and identified LRP2 mutations in six families with Donnai-Barrow syndrome and one family with facio-oculo-acoustico-renal syndrome. LRP2 encodes megalin, a multiligand uptake receptor that regulates levels of diverse circulating compounds. This work implicates a pathway with potential pharmacological therapeutic targets.


American Journal of Medical Genetics Part A | 2006

Findings From aCGH in Patients With Congenital Diaphragmatic Hernia (CDH): A Possible Locus for Fryns Syndrome

Sibel Kantarci; David Casavant; C. Prada; Meaghan K Russell; Janice L. B. Byrne; L. Wilkins Haug; Russell W. Jennings; Simon M. Manning; Theonia K. Boyd; Jean Pierre Fryns; Lewis B. Holmes; Patricia K. Donahoe; Charles Lee; Virginia E. Kimonis; Barbara R. Pober

Congenital diaphragmatic hernia (CDH) is a common and often devastating birth defect that can occur in isolation or as part of a malformation complex. Considerable progress is being made in the identification of genetic causes of CDH. We applied array‐based comparative genomic hybridization (aCGH) of ∼1Mb resolution to 29 CDH patients with prior normal karyotypes who had been recruited into our multi‐site study. One patient, clinically diagnosed with Fryns syndrome, demonstrated a de novo 5Mb deletion at chromosome region 1q41–q42.12 that was confirmed by FISH. Given prior reports of CDH in association with cytogenetic abnormalities in this region, we propose that this represents a locus for Fryns syndrome, a Fryns syndrome phenocopy, or CDH.


American Journal of Medical Genetics Part A | 2005

Infants with Bochdalek diaphragmatic hernia: Sibling precurrence and monozygotic twin discordance in a hospital‐based malformation surveillance program

Barbara R. Pober; Angela E. Lin; Meaghan K Russell; Kate G. Ackerman; Sharmila Chakravorty; Bernarda Strauss; Marie Noel Westgate; Jay M. Wilson; Patricia K. Donahoe; Lewis B. Holmes

Congenital diaphragmatic hernia (CDH) is a common and often devastating birth defect. In order to learn more about possible genetic causes, we reviewed and classified 203 cases of the Bochdalek hernia type identified through the Brigham and Womens Hospital (BWH) Active Malformation Surveillance Program over a 28‐year period. Phenotypically, 55% of the cases had isolated CDH, and 45% had complex CDH defined as CDH in association with additional major malformations or as part of a syndrome. When classified according to likely etiology, 17% had a Recognized Genetic etiology for their CDH, while the remaining 83% had No Apparent Genetic etiology. Detailed analysis using this largest cohort of consecutively collected cases of CDH showed low precurrence among siblings. Additionally, there was no concordance for CDH among five monozygotic twin pairs. These findings, in conjunction with previous reports of de novo dominant mutations in patients with CDH, suggest that new mutations may be an important mechanism responsible for CDH. The twin data also raise the possibility that epigenetic abnormalities contribute to the development of CDH.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Congenital diaphragmatic hernia candidate genes derived from embryonic transcriptomes

Meaghan K Russell; Mauro Longoni; Julie Wells; Faouzi I. Maalouf; Adam A. Tracy; Maria Loscertales; Kate G. Ackerman; Barbara R. Pober; Kasper Lage; Patricia K. Donahoe

Congenital diaphragmatic hernia (CDH) is a common (1 in 3,000 live births) major congenital malformation that results in significant morbidity and mortality. The discovery of CDH loci using standard genetic approaches has been hindered by its genetic heterogeneity. We hypothesized that gene expression profiling of developing embryonic diaphragms would help identify genes likely to be associated with diaphragm defects. We generated a time series of whole-transcriptome expression profiles from laser captured embryonic mouse diaphragms at embryonic day (E)11.5 and E12.5 when experimental perturbations lead to CDH phenotypes, and E16.5 when the diaphragm is fully formed. Gene sets defining biologically relevant pathways and temporal expression trends were identified by using a series of bioinformatic algorithms. These developmental sets were then compared with a manually curated list of genes previously shown to cause diaphragm defects in humans and in mouse models. Our integrative filtering strategy identified 27 candidates for CDH. We examined the diaphragms of knockout mice for one of the candidate genes, pre–B-cell leukemia transcription factor 1 (Pbx1), and identified a range of previously undetected diaphragmatic defects. Our study demonstrates the utility of genetic characterization of normal development as an integral part of a disease gene identification and prioritization strategy for CDH, an approach that can be extended to other diseases and developmental anomalies.


American Journal of Medical Genetics Part A | 2008

Donnai–Barrow syndrome (DBS/FOAR) in a child with a homozygous LRP2 mutation due to complete chromosome 2 paternal isodisomy†‡

Sibel Kantarci; Nicola Ragge; N. Simon Thomas; David O. Robinson; Kristin M Noonan; Meaghan K Russell; Dian Donnai; F. Lucy Raymond; Christopher A. Walsh; Patricia K. Donahoe; Barbara R. Pober

Donnai–Barrow syndrome [Faciooculoacousticorenal (FOAR) syndrome; DBS/FOAR] is a rare autosomal recessive disorder resulting from mutations in the LRP2 gene located on chromosome 2q31.1. We report a unique DBS/FOAR patient homozygous for a 4‐bp LRP2 deletion secondary to paternal uniparental isodisomy for chromosome 2. The propositus inherited the mutation from his heterozygous carrier father, whereas the mother carried only wild‐type LRP2 alleles. This is the first case of DBS/FOAR resulting from uniparental disomy (UPD) and the fourth published case of any paternal UPD 2 ascertained through unmasking of an autosomal recessive disorder. The absence of clinical symptoms above and beyond the classical phenotype in this and the other disorders suggests that paternal chromosome 2 is unlikely to contain imprinted genes notably affecting either growth or development. This report highlights the importance of parental genotyping in order to give accurate genetic counseling for autosomal recessive disorders.


American Journal of Medical Genetics Part A | 2012

Congenital diaphragmatic hernia interval on chromosome 8p23.1 characterized by genetics and protein interaction networks

Mauro Longoni; Kasper Lage; Meaghan K Russell; Maria Loscertales; Omar A. Abdul-Rahman; Gareth Baynam; Steven B. Bleyl; Paul Brady; Jeroen Breckpot; Chih P. Chen; Koenraad Devriendt; Gabriele Gillessen-Kaesbach; Arthur W. Grix; Alan F. Rope; Osamu Shimokawa; Bernarda Strauss; Dagmar Wieczorek; Elaine H. Zackai; Caroline Coletti; Faouzi I. Maalouf; Kristin M Noonan; Ji H. Park; Adam A. Tracy; Charles Lee; Patricia K. Donahoe; Barbara R. Pober

Chromosome 8p23.1 is a common hotspot associated with major congenital malformations, including congenital diaphragmatic hernia (CDH) and cardiac defects. We present findings from high‐resolution arrays in patients who carry a loss (n = 18) or a gain (n = 1) of sub‐band 8p23.1. We confirm a region involved in both diaphragmatic and heart malformations. Results from a novel CNVConnect algorithm, prioritizing protein–protein interactions between products of genes in the 8p23.1 hotspot and products of previously known CDH causing genes, implicated GATA4, NEIL2, and SOX7 in diaphragmatic defects. Sequence analysis of these genes in 226 chromosomally normal CDH patients, as well as in a small number of deletion 8p23.1 patients, showed rare unreported variants in the coding region; these may be contributing to the diaphragmatic phenotype. We also demonstrated that two of these three genes were expressed in the E11.5–12.5 primordial mouse diaphragm, the developmental stage at which CDH is thought to occur. This combination of bioinformatics and expression studies can be applied to other chromosomal hotspots, as well as private microdeletions or microduplications, to identify causative genes and their interaction networks.


Clinical Genetics | 2015

Prevalence and penetrance of ZFPM2 mutations and deletions causing congenital diaphragmatic hernia.

Mauro Longoni; Meaghan K Russell; Frances A. High; Katayoon Darvishi; Faouzi I. Maalouf; Alireza Kashani; Adam A. Tracy; Caroline Coletti; Maria Loscertales; Kasper Lage; Kate G. Ackerman; S A Woods; C Ward-Melver; D Andrews; Charles Lee; Barbara R. Pober; Patricia K. Donahoe

Zinc finger protein, FOG2 family member 2 (ZFPM2) (previously named FOG2) gene defects result in the highly morbid congenital diaphragmatic hernia (CDH) in humans and animal models. In a cohort of 275 CDH patient exomes, we estimated the prevalence of damaging ZFPM2 mutations to be almost 5%. Genetic analysis of a multigenerational family identified a heritable intragenic ZFPM2 deletion with an estimated penetrance of 37.5%, which has important implications for genetic counseling. Similarly, a low penetrance ZFPM2 frameshift mutation was observed in a second multiplex family. Isolated CDH was the predominant phenotype observed in our ZFPM2 mutation patients. Findings from the patients described herein indicate that ZFPM2 point mutations or deletions are a recurring cause of CDH.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Molecular pathogenesis of congenital diaphragmatic hernia revealed by exome sequencing, developmental data, and bioinformatics

Mauro Longoni; Frances A. High; Meaghan K Russell; Alireza Kashani; Adam A. Tracy; Caroline Coletti; Regis Hila; Ahmed Shamia; Julie Wells; Kate G. Ackerman; Jay M. Wilson; Charles C. Lee; Kasper Lage; Barbara R. Pober; Patricia K. Donahoe

Significance Congenital diaphragmatic hernia (CDH) is a common birth defect associated with high morbidity and mortality. Focusing on the coding sequence of 51 genes, discovered in human studies and in mouse models, we studied 275 CDH patients and identified multiple variants in CDH-causing genes. Information on gene expression in embryonic mouse diaphragms and protein interactions allowed us to prioritize additional compelling CDH-associated genes. We believe that an improved understanding of the genetics of CDH will be important to design new therapeutic strategies for patients with diaphragmatic defects. Congenital diaphragmatic hernia (CDH) is a common and severe birth defect. Despite its clinical significance, the genetic and developmental pathways underlying this disorder are incompletely understood. In this study, we report a catalog of variants detected by a whole exome sequencing study on 275 individuals with CDH. Predicted pathogenic variants in genes previously identified in either humans or mice with diaphragm defects are enriched in our CDH cohort compared with 120 size-matched random gene sets. This enrichment was absent in control populations. Variants in these critical genes can be found in up to 30.9% of individuals with CDH. In addition, we filtered variants by using genes derived from regions of recurrent copy number variations in CDH, expression profiles of the developing diaphragm, protein interaction networks expanded from the known CDH-causing genes, and prioritized genes with ultrarare and highly disruptive variants, in 11.3% of CDH patients. These strategies have identified several high priority genes and developmental pathways that likely contribute to the CDH phenotype. These data are valuable for comparison of candidate genes generated from whole exome sequencing of other CDH cohorts or multiplex kindreds and provide ideal candidates for further functional studies. Furthermore, we propose that these genes and pathways will enhance our understanding of the heterogeneous molecular etiology of CDH.


Clinical Dysmorphology | 2010

Focal segmental glomerulosclerosis in a female patient with Donnai–barrow syndrome

Ihab Sakr Shaheen; Eric Finlay; Katrina Prescott; Meaghan K Russell; Mauro Longoni; Shelagh Joss

Case report The patient was born by normal vaginal delivery at 38 weeks to consanguineous parents. Her mother had had regular antenatal follow-up, and a scan at 36 weeks gestation showed isolated agenesis of the corpus callosum. Her birth weight was 2810 g (50th centile) and her head circumference was 34.5 cm (91st centile). She was noted to have bilateral iris colobomata, downslanting palpebral fissures, hypertelorism, a broad nasal root and flattened nasal tip, a wide philtrum and low-set ears (Fig. 1).


Proceedings of the National Academy of Sciences of the United States of America | 2018

Systematic analysis of copy number variation associated with congenital diaphragmatic hernia

Qihui Zhu; Frances A. High; Chengsheng Zhang; Eliza Cerveira; Meaghan K Russell; Mauro Longoni; Maliackal Poulo Joy; Mallory Ryan; Adam Mil-homens; Lauren Bellfy; Caroline Coletti; Pooja Bhayani; Regis Hila; Jay M. Wilson; Patricia K. Donahoe; Charles Lee

Significance This study describes the results of a large-scale case control analysis of copy number variants (CNVs) in a cohort of patients with congenital diaphragmatic hernia (CDH) and a large number of healthy population-matched controls. Using a customized array comparative genomic hybridization system, we have identified six CNVs that are associated with CDH with statistical significance (P < 0.05). These regions validate several hypothesized CDH candidate genes and identify additional genes and pathways that contribute to the pathogenesis of CDH. The estimated frequency of pathogenic CNVs in this cohort is 13%, which underscores the critical contribution of CNVs in CDH. This study also provides a model approach that is broadly applicable to other structural birth defects and identifies candidates for future functional studies. Congenital diaphragmatic hernia (CDH), characterized by malformation of the diaphragm and hypoplasia of the lungs, is one of the most common and severe birth defects, and is associated with high morbidity and mortality rates. There is growing evidence demonstrating that genetic factors contribute to CDH, although the pathogenesis remains largely elusive. Single-nucleotide polymorphisms have been studied in recent whole-exome sequencing efforts, but larger copy number variants (CNVs) have not yet been studied on a large scale in a case control study. To capture CNVs within CDH candidate regions, we developed and tested a targeted array comparative genomic hybridization platform to identify CNVs within 140 regions in 196 patients and 987 healthy controls, and identified six significant CNVs that were either unique to patients or enriched in patients compared with controls. These CDH-associated CNVs reveal high-priority candidate genes including HLX, LHX1, and HNF1B. We also discuss CNVs that are present in only one patient in the cohort but have additional evidence of pathogenicity, including extremely rare large and/or de novo CNVs. The candidate genes within these predicted disease-causing CNVs form functional networks with other known CDH genes and play putative roles in DNA binding/transcription regulation and embryonic development. These data substantiate the importance of CNVs in the etiology of CDH, identify CDH candidate genes and pathways, and highlight the importance of ongoing analysis of CNVs in the study of CDH and other structural birth defects.

Collaboration


Dive into the Meaghan K Russell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles Lee

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jay M. Wilson

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge