Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
EBioMedicine ; 71: 103564, 2021 09.
Article in English | MEDLINE | ID: mdl-34455395

Subject(s)
Hernia, Inguinal , Humans
2.
Hum Mol Genet ; 29(22): 3606-3615, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33179741

ABSTRACT

Adolescent idiopathic scoliosis (AIS), a sideways curvature of the spine, is the most common pediatric musculoskeletal disorder, affecting ~3% of the population worldwide. However, its genetic bases and tissues of origin remain largely unknown. Several genome-wide association studies (GWAS) have implicated nucleotide variants in non-coding sequences that control genes with important roles in cartilage, muscle, bone, connective tissue and intervertebral disks (IVDs) as drivers of AIS susceptibility. Here, we set out to define the expression of AIS-associated genes and active regulatory elements by performing RNA-seq and chromatin immunoprecipitation-sequencing against H3 lysine 27 acetylation in these tissues in mouse and human. Our study highlights genetic pathways involving AIS-associated loci that regulate chondrogenesis, IVD development and connective tissue maintenance and homeostasis. In addition, we identify thousands of putative AIS-associated regulatory elements which may orchestrate tissue-specific expression in musculoskeletal tissues of the spine. Quantification of enhancer activity of several candidate regulatory elements from our study identifies three functional enhancers carrying AIS-associated GWAS SNPs at the ADGRG6 and BNC2 loci. Our findings provide a novel genome-wide catalog of AIS-relevant genes and regulatory elements and aid in the identification of novel targets for AIS causality and treatment.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Histones/genetics , Receptors, G-Protein-Coupled/genetics , Scoliosis/genetics , Acetylation , Adolescent , Child , Female , Genome-Wide Association Study , Genomics/trends , Humans , Lysine/genetics , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , RNA-Seq , Scoliosis/epidemiology , Scoliosis/pathology , Spine/metabolism , Spine/pathology , Transcriptome/genetics
3.
Bone Res ; 8: 13, 2020.
Article in English | MEDLINE | ID: mdl-32195011

ABSTRACT

The human spinal column is a dynamic, segmented, bony, and cartilaginous structure that protects the neurologic system and simultaneously provides balance and flexibility. Children with developmental disorders that affect the patterning or shape of the spine can be at risk of neurologic and other physiologic dysfunctions. The most common developmental disorder of the spine is scoliosis, a lateral deformity in the shape of the spinal column. Scoliosis may be part of the clinical spectrum that is observed in many developmental disorders, but typically presents as an isolated symptom in otherwise healthy adolescent children. Adolescent idiopathic scoliosis (AIS) has defied understanding in part due to its genetic complexity. Breakthroughs have come from recent genome-wide association studies (GWAS) and next generation sequencing (NGS) of human AIS cohorts, as well as investigations of animal models. These studies have identified genetic associations with determinants of cartilage biogenesis and development of the intervertebral disc (IVD). Current evidence suggests that a fraction of AIS cases may arise from variation in factors involved in the structural integrity and homeostasis of the cartilaginous extracellular matrix (ECM). Here, we review the development of the spine and spinal cartilages, the composition of the cartilage ECM, the so-called "matrisome" and its functions, and the players involved in the genetic architecture of AIS. We also propose a molecular model by which the cartilage matrisome of the IVD contributes to AIS susceptibility.

4.
PLoS Genet ; 15(10): e1008096, 2019 10.
Article in English | MEDLINE | ID: mdl-31652254

ABSTRACT

Degenerative changes of the intervertebral disc (IVD) are a leading cause of disability affecting humans worldwide and has been attributed primarily to trauma and the accumulation of pathology during aging. While genetic defects have also been associated with disc degeneration, the precise mechanisms driving the initiation and progression of disease have remained elusive due to a paucity of genetic animal models. Here, we discuss a novel conditional mouse genetic model of endplate-oriented disc herniations in adult mice. Using conditional mouse genetics, we show increased mechanical stiffness and reveal dysregulation of typical gene expression profiles of the IVD in adhesion G-protein coupled receptor G6 (Adgrg6) mutant mice prior to the onset of endplate-oriented disc herniations in adult mice. We observed increased STAT3 activation prior to IVD defects and go on to demonstrate that treatment of Adgrg6 conditional mutant mice with a small molecule inhibitor of STAT3 activation ameliorates endplate-oriented herniations. These findings establish ADGRG6 and STAT3 as novel regulators of IVD endplate and growth plate integrity in the mouse, and implicate ADGRG6/STAT3 signaling as promising therapeutic targets for endplate-oriented disc degeneration.


Subject(s)
Intervertebral Disc Degeneration/genetics , Intervertebral Disc Displacement/genetics , Receptors, G-Protein-Coupled/genetics , STAT3 Transcription Factor/genetics , Animals , Disease Models, Animal , Disease Progression , Growth Plate , Humans , Intervertebral Disc/growth & development , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/physiopathology , Intervertebral Disc Displacement/physiopathology , Mice , Mutation , Signal Transduction
5.
Hum Mol Genet ; 27(22): 3986-3998, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30395268

ABSTRACT

Adolescent idiopathic scoliosis (AIS) is the most common musculoskeletal disorder of childhood development. The genetic architecture of AIS is complex, and the great majority of risk factors are undiscovered. To identify new AIS susceptibility loci, we conducted the first genome-wide meta-analysis of AIS genome-wide association studies, including 7956 cases and 88 459 controls from 3 ancestral groups. Three novel loci that surpassed genome-wide significance were uncovered in intragenic regions of the CDH13 (P-value_rs4513093 = 1.7E-15), ABO (P-value_ rs687621 = 7.3E-10) and SOX6 (P-value_rs1455114 = 2.98E-08) genes. Restricting the analysis to females improved the associations at multiple loci, most notably with variants within CDH13 despite the reduction in sample size. Genome-wide gene-functional enrichment analysis identified significant perturbation of pathways involving cartilage and connective tissue development. Expression of both SOX6 and CDH13 was detected in cartilage chondrocytes and chromatin immunoprecipitation sequencing experiments in that tissue revealed multiple HeK27ac-positive peaks overlapping associated loci. Our results further define the genetic architecture of AIS and highlight the importance of vertebral cartilage development in its pathogenesis.


Subject(s)
ABO Blood-Group System/genetics , Cadherins/genetics , Musculoskeletal Diseases/genetics , SOXD Transcription Factors/genetics , Scoliosis/genetics , Adolescent , Child , Ethnicity/genetics , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Musculoskeletal Diseases/physiopathology , Polymorphism, Single Nucleotide/genetics , Scoliosis/physiopathology , Young Adult
6.
Anat Rec (Hoboken) ; 300(10): 1838-1846, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28667681

ABSTRACT

The rhombomeric compartments of the hindbrain are characterized by lineage restriction; cells born in one compartment generally remain there and do not migrate to neighboring rhombomeres. Two well-known exceptions are the substantial migrations of the pontine nuclei and the mammalian facial nucleus. In this study we used Hoxa3-Cre lineage to permanently mark cells that originate in rhombomeres caudal to r4. We found that cells born caudal to the r4/r5 border migrate forwards to a number of different locations in rhombomeres 1-4; the final locations include the interfascicular trigeminal nucleus, the principal trigeminal nucleus, the pontine nuclei, the reticulotegmental nucleus, the ventral nucleus of the lateral lemniscus, and the lateral and medial vestibular nuclei. We suggest that there are numerous exceptions to the principle of rhombomeric lineage restriction that have previously gone unnoticed. Anat Rec, 2017. © 2017 Wiley Periodicals, Inc. Anat Rec, 300:1838-1846, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Rhombencephalon/cytology , Rhombencephalon/embryology , Animals , Cell Lineage , Cell Movement , Homeodomain Proteins/analysis , Mice, Transgenic
7.
Nat Genet ; 48(5): 528-36, 2016 05.
Article in English | MEDLINE | ID: mdl-27019111

ABSTRACT

Bats are the only mammals capable of powered flight, but little is known about the genetic determinants that shape their wings. Here we generated a genome for Miniopterus natalensis and performed RNA-seq and ChIP-seq (H3K27ac and H3K27me3) analyses on its developing forelimb and hindlimb autopods at sequential embryonic stages to decipher the molecular events that underlie bat wing development. Over 7,000 genes and several long noncoding RNAs, including Tbx5-as1 and Hottip, were differentially expressed between forelimb and hindlimb, and across different stages. ChIP-seq analysis identified thousands of regions that are differentially modified in forelimb and hindlimb. Comparative genomics found 2,796 bat-accelerated regions within H3K27ac peaks, several of which cluster near limb-associated genes. Pathway analyses highlighted multiple ribosomal proteins and known limb patterning signaling pathways as differentially regulated and implicated increased forelimb mesenchymal condensation in differential growth. In combination, our work outlines multiple genetic components that likely contribute to bat wing formation, providing insights into this morphological innovation.


Subject(s)
Chiroptera/embryology , Chiroptera/genetics , Epigenesis, Genetic , Transcriptome , Wings, Animal/embryology , Animals , Embryonic Development/genetics , Gene Expression Profiling , Genome , Male , RNA, Long Noncoding , Regulatory Sequences, Nucleic Acid , Sequence Analysis, RNA
8.
Nat Commun ; 6: 10130, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26686553

ABSTRACT

Inguinal hernia repair is one of the most commonly performed operations in the world, yet little is known about the genetic mechanisms that predispose individuals to develop inguinal hernias. We perform a genome-wide association analysis of surgically confirmed inguinal hernias in 72,805 subjects (5,295 cases and 67,510 controls) and confirm top associations in an independent cohort of 92,444 subjects with self-reported hernia repair surgeries (9,701 cases and 82,743 controls). We identify four novel inguinal hernia susceptibility loci in the regions of EFEMP1, WT1, EBF2 and ADAMTS6. Moreover, we observe expression of all four genes in mouse connective tissue and network analyses show an important role for two of these genes (EFEMP1 and WT1) in connective tissue maintenance/homoeostasis. Our findings provide insight into the aetiology of hernia development and highlight genetic pathways for studies of hernia development and its treatment.


Subject(s)
ADAM Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Extracellular Matrix Proteins/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Hernia, Inguinal/genetics , WT1 Proteins/genetics , ADAMTS Proteins , Adult , Aged , Aged, 80 and over , Animals , Cohort Studies , Female , Humans , Male , Mice , Middle Aged
9.
J Comp Neurol ; 521(3): 697-708, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-22829396

ABSTRACT

We have found a previously unreported precerebellar nucleus located among the emerging fibers of the motor root of the trigeminal nerve in the mouse, which we have called the interfascicular trigeminal nucleus (IF5). This nucleus had previously been named the tensor tympani part of the motor trigeminal nucleus (5TT) in rodent brain atlases, because it was thought to be a subset of small motor neurons of the motor trigeminal nucleus innervating the tensor tympani muscle. However, following injection of retrograde tracer in the cerebellum, the labeled neurons in IF5 were found to be choline acetyltransferase (ChAT) negative, indicating that they are not motor neurons. The cells of IF5 are strongly labeled in mice from Wnt1Cre and Atoh1 CreER lineage fate mapping, in common with the major precerebellar nuclei that arise from the rhombic lip and that issue mossy fibers. Analysis of sections from mouse Hoxa3, Hoxb1, and Egr2 Cre labeled lineages shows that the neurons of IF5 arise from rhombomeres caudal to rhombomere 4, most likely from rhombomeres 6-8. We conclude that IF5 is a significant precerebellar nucleus in the mouse that shares developmental gene expression characteristics with mossy fiber precerebellar nuclei that arise from the caudal rhombic lip.


Subject(s)
Efferent Pathways/cytology , Efferent Pathways/embryology , Pons/cytology , Pons/embryology , Trigeminal Nuclei/cytology , Trigeminal Nuclei/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Lineage/genetics , Cerebellum/cytology , Cerebellum/embryology , Cerebellum/physiology , Choline O-Acetyltransferase/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Efferent Pathways/physiology , Female , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Fibers/physiology , Neuronal Tract-Tracers , Pons/physiology , Trigeminal Nuclei/physiology , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
10.
Hum Mol Genet ; 21(1): 26-31, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21940751

ABSTRACT

Congenital heart disease is one of the most common human birth defects, yet many genes and pathways regulating heart development remain unknown. A recent study in humans revealed that mutations in a single Hox gene, HOXA1 (Athabascan Brainstem Dysgenesis Syndrome, Bosley-Salih-Alorainy Syndrome), can cause severe cardiovascular malformations, some of which are lethal without surgical intervention. Since the discovery of the human syndromes, there have been no reports of any Hox mouse mutants with cardiac defects, hampering studies to explore the developmental causes of the human disease. In this study, we identify severe cardiovascular malformations in a Hox mouse model, which mimic the congenital heart defects in HOXA1 syndrome patients. Hoxa1 null mice show defects such as interrupted aortic arch, aberrant subclavian artery and Tetralogy of Fallot, demonstrating that Hoxa1 is required for patterning of the great arteries and outflow tract of the heart. We show that during early embryogenesis, Hoxa1 is expressed in precursors of cardiac neural crest cells (NCCs), which populate the heart. We further demonstrate that Hoxa1 acts upstream of several genes, important for neural crest specification. Thus, our data allow us to suggest a model in which Hoxa1 regulates heart development through its influence on cardiac NCCs, providing insight into the mechanisms underlying the human disease.


Subject(s)
Disease Models, Animal , Heart Defects, Congenital/genetics , Homeodomain Proteins/genetics , Mice , Transcription Factors/genetics , Animals , Base Sequence , Female , Heart/growth & development , Heart/physiopathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/physiopathology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Phenotype , Transcription Factors/deficiency
11.
Dev Biol ; 357(2): 295-304, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21784065

ABSTRACT

Hox genes play a crucial role during embryonic patterning and organogenesis. Of the 39 Hox genes, Hoxa1 is the first to be expressed during embryogenesis and the only anterior Hox gene linked to a human syndrome. Hoxa1 is necessary for the proper development of the brainstem, inner ear and heart in humans and mice; however, almost nothing is known about the molecular downstream targets through which it exerts its function. To gain insight into the transcriptional network regulated by this protein, we performed microarray analysis on tissue microdissected from the prospective rhombomere 3-5 region of Hoxa1 null and wild type embryos. Due to the very early and transient expression of this gene, dissections were performed on early somite stage embryos during an eight-hour time window of development. Our array yielded a list of around 300 genes differentially expressed between the two samples. Many of the identified genes play a role in a specific developmental or cellular process. Some of the validated targets regulate early neural crest induction and specification. Interestingly, three of these genes, Zic1, Hnf1b and Foxd3, were down-regulated in the posterior hindbrain, where cardiac neural crest cells arise, which pattern the outflow tract of the heart. Other targets are necessary for early inner ear development, e.g. Pax8 and Fgfr3 or are expressed in specific hindbrain neurons regulating respiration, e.g. Lhx5. These findings allow us to propose a model where Hoxa1 acts in a genetic cascade upstream of genes controlling specific aspects of embryonic development, thereby providing insight into possible mechanisms underlying the human HoxA1-syndrome.


Subject(s)
Ear, Inner/embryology , Homeodomain Proteins/metabolism , Neural Crest/embryology , Rhombencephalon/embryology , Transcription Factors/metabolism , Animals , Ear, Inner/cytology , Ear, Inner/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Gene Targeting , Homeodomain Proteins/genetics , Humans , In Situ Hybridization , Mice , Mice, Inbred C57BL , Models, Biological , Neural Crest/cytology , Neural Crest/metabolism , Oligonucleotide Array Sequence Analysis , Organ Specificity/genetics , Phenotype , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Rhombencephalon/cytology , Rhombencephalon/metabolism , Time Factors , Transcription Factors/genetics
12.
Cerebellum ; 10(3): 570-84, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21479970

ABSTRACT

The precerebellar nuclei are hindbrain and spinal cord centers that send fibers to the cerebellum. The neurons of the major hindbrain precerebellar nuclei are derived from the rhombic lip. Wnt1, a developmentally important gene involved in intercellular signaling, is expressed in the developing rhombic lip. We sought to investigate the relationship between the cell clusters expressing Wnt1 and the precerebellar nuclei in the hindbrain. We therefore defined the hindbrain precerebellar nuclei by retrograde tracing, following cerebellar injections of HRP, and compared these results with the cell clusters expressing Wnt1 in newborn mice. We found that 39 distinct hindbrain nuclei project to the cerebellum. Of these nuclei, all but three (namely the oral pontine reticular nucleus, the caudal pontine reticular nucleus, and the subcoeruleus nucleus) contain neurons expressing Wnt1. This shows a high degree of overlap between the precerebellar nuclei and the nuclei that express Wnt1. However, it should be noted that neurons expressing Wnt1 are also found in the superior olivary complex, which is a basal plate derivative lacking cerebellar projections.


Subject(s)
Brain Mapping , Cerebellum/physiology , Neurons/physiology , Rhombencephalon/cytology , Wnt1 Protein/genetics , Amino Acids/metabolism , Animals , Functional Laterality , Galactosides/genetics , Galactosides/metabolism , Horseradish Peroxidase/metabolism , Indoles/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/physiology , Proteins/genetics , Proteins/metabolism , RNA, Untranslated , Stilbamidines/metabolism , Wnt1 Protein/metabolism
13.
Dev Biol ; 341(2): 499-509, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20171203

ABSTRACT

Loss of Hoxa1 function results in severe defects of the brainstem, inner ear, and cranial ganglia in humans and mice as well as cardiovascular abnormalities in humans. Because Hoxa1 is expressed very transiently during an early embryonic stage, it has been difficult to determine whether Hoxa1 plays a direct role in the precursors of the affected organs or if all defects result from indirect effects due to mispatterning of the hindbrain. In this study we use a Hoxa1-IRES-Cre mouse to genetically label the early Hoxa1-expressing cells and determine their contribution to each of the affected organs, allowing us to conclude in which precursor tissue Hoxa1 is expressed. We found Hoxa1 lineage-labeled cells in all tissues expected to be derived from the Hoxa1 domain, such as the facial and abducens nuclei and nerves as well as r4 neural crest cells. In addition, we detected the lineage in derivatives that were not thought to have expressed Hoxa1 during development. In the brainstem, the anterior border of the lineage was found to be in r3, which is more anterior than previously reported. We also observed an interesting pattern of the lineage in the inner ear, namely a strong contribution to the otic epithelium with the exception of sensory patches. Moreover, lineage-labeled cells were detected in the atria and outflow tract of the developing heart. In conclusion, Hoxa1 lineage tracing uncovered new domains of Hoxa1 expression in rhombomere 3, the otic epithelium, and cardiac precursors, suggesting a more direct role for Hoxa1 in development of these tissues than previously believed.


Subject(s)
Brain Stem/embryology , Ear, Inner/embryology , Heart/embryology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Mice
14.
Brain Res ; 1271: 49-59, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19281800

ABSTRACT

The linear nucleus (Li) is a prominent cell group in the caudal hindbrain, which was first described in a study of cerebellar afferents in the rat by [Watson, C.R.R., Switzer, R.C. III, 1978. Trigeminal projections to cerebellar tactile areas in the rat origin mainly from N. interpolaris and N. principalis. Neurosci. Lett. 10, 77-82.]. It was named for its elongated appearance in transverse sections. Since this original description in the rat, reference to the nucleus seems to have been largely absent from experimental studies of mammalian precerebellar nuclei. We therefore set out to define the cytoarchitecture, cerebellar connections, and molecular characteristics of Li in the mouse. In coronal Nissl sections at the level of the rostral inferior olive, it consists of two parallel bands of cells joined at their dorsal apex by a further band of cells, making the shape of the Greek capital letter pi. Our three-dimensional reconstruction demonstrated that the nucleus is continuous with the lateral reticular nucleus (LRt) and that the ambiguus nucleus sits inside the arch of Li. Cerebellar horseradish peroxidase injections confirmed that the cells of Li project to cerebellum. We have shown that Li cells express Atoh1 and Wnt1 lineage markers that are known to label the rhombic lip derived precerebellar nuclei. We have examined the relationship of Li cells to a number of molecular markers, and have found that many of the cells express a nonphosphorylated epitope in neurofilament H (SMI 32), a feature they share with the LRt. The mouse Li therefore appears to be a rostrodorsal extension of the LRt.


Subject(s)
Cerebellum/cytology , Cerebellum/metabolism , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Reticular Formation/cytology , Reticular Formation/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/analysis , Brain Mapping , Cell Lineage/physiology , Cerebellum/embryology , Galactosides , Gene Expression , Genes, Reporter , Horseradish Peroxidase , Image Processing, Computer-Assisted , Indoles , Medulla Oblongata/embryology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/metabolism , Neurofilament Proteins/analysis , Neurofilament Proteins/genetics , Reticular Formation/embryology , Rhombencephalon/cytology , Rhombencephalon/embryology , Rhombencephalon/metabolism , Staining and Labeling , Wnt1 Protein/analysis , Wnt1 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...