Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
JCI Insight ; 8(21)2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37796615

ABSTRACT

Osteogenesis imperfecta (OI), or brittle bone disease, is a disorder characterized by bone fragility and increased fracture incidence. All forms of OI also feature short stature, implying an effect on endochondral ossification. Using the Aga2+/- mouse, which has a mutation in type I collagen, we show an affected growth plate primarily due to a shortened proliferative zone. We used single-cell RNA-Seq analysis of tibial and femoral growth plate tissues to understand transcriptional consequences on growth plate cell types. We show that perichondrial cells, which express abundant type I procollagen, and growth plate chondrocytes, which were found to express low amounts of type I procollagen, had ER stress and dysregulation of the same unfolded protein response pathway as previously demonstrated in osteoblasts. Aga2+/- proliferating chondrocytes showed increased FGF and MAPK signaling, findings consistent with accelerated differentiation. There was also increased Sox9 expression throughout the growth plate, which is expected to accelerate early chondrocyte differentiation but reduce late hypertrophic differentiation. These data reveal that mutant type I collagen expression in OI has an impact on the cartilage growth plate. These effects on endochondral ossification indicate that OI is a biologically complex phenotype going beyond its known impacts on bone to negatively affect linear growth.


Subject(s)
Osteogenesis Imperfecta , Animals , Mice , Cartilage/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Gene Expression , Osteogenesis Imperfecta/metabolism
2.
Bone Res ; 10(1): 37, 2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35474298

ABSTRACT

Spondylocarpotarsal syndrome (SCT) is a rare musculoskeletal disorder characterized by short stature and vertebral, carpal, and tarsal fusions resulting from biallelic nonsense mutations in the gene encoding filamin B (FLNB). Utilizing a FLNB knockout mouse, we showed that the vertebral fusions in SCT evolved from intervertebral disc (IVD) degeneration and ossification of the annulus fibrosus (AF), eventually leading to full trabecular bone formation. This resulted from alterations in the TGFß/BMP signaling pathway that included increased canonical TGFß and noncanonical BMP signaling. In this study, the role of FLNB in the TGFß/BMP pathway was elucidated using in vitro, in vivo, and ex vivo treatment methodologies. The data demonstrated that FLNB interacts with inhibitory Smads 6 and 7 (i-Smads) to regulate TGFß/BMP signaling and that loss of FLNB produces increased TGFß receptor activity and decreased Smad 1 ubiquitination. Through the use of small molecule inhibitors in an ex vivo spine model, TGFß/BMP signaling was modulated to design a targeted treatment for SCT and disc degeneration. Inhibition of canonical and noncanonical TGFß/BMP pathway activity restored Flnb-/- IVD morphology. These most effective improvements resulted from specific inhibition of TGFß and p38 signaling activation. FLNB acts as a bridge for TGFß/BMP signaling crosstalk through i-Smads and is key for the critical balance in TGFß/BMP signaling that maintains the IVD. These findings further our understanding of IVD biology and reveal new molecular targets for disc degeneration as well as congenital vertebral fusion disorders.

3.
J Bone Miner Res ; 37(4): 675-686, 2022 04.
Article in English | MEDLINE | ID: mdl-34997935

ABSTRACT

Osteogenesis imperfecta (OI) is a genetically heterogenous disorder most often due to heterozygosity for mutations in the type I procollagen genes, COL1A1 or COL1A2. The disorder is characterized by bone fragility leading to increased fracture incidence and long-bone deformities. Although multiple mechanisms underlie OI, endoplasmic reticulum (ER) stress as a cellular response to defective collagen trafficking is emerging as a contributor to OI pathogenesis. Herein, we used 4-phenylbutiric acid (4-PBA), an established chemical chaperone, to determine if treatment of Aga2+/- mice, a model for moderately severe OI due to a Col1a1 structural mutation, could attenuate the phenotype. In vitro, Aga2+/- osteoblasts show increased protein kinase RNA-like endoplasmic reticulum kinase (PERK) activation protein levels, which improved upon treatment with 4-PBA. The in vivo data demonstrate that a postweaning 5-week 4-PBA treatment increased total body length and weight, decreased fracture incidence, increased femoral bone volume fraction (BV/TV), and increased cortical thickness. These findings were associated with in vivo evidence of decreased bone-derived protein levels of the ER stress markers binding immunoglobulin protein (BiP), CCAAT/-enhancer-binding protein homologous protein (CHOP), and activating transcription factor 4 (ATF4) as well as increased levels of the autophagosome marker light chain 3A/B (LC3A/B). Genetic ablation of CHOP in Aga2+/- mice resulted in increased severity of the Aga2+/- phenotype, suggesting that the reduction in CHOP observed in vitro after treatment is a consequence rather than a cause of reduced ER stress. These findings suggest the potential use of chemical chaperones as an adjunct treatment for forms of OI associated with ER stress. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Osteogenesis Imperfecta , Animals , Butylamines , Collagen Type I/metabolism , Disease Models, Animal , Mice , Molecular Chaperones/metabolism , Mutation , Osteoblasts/metabolism , Osteogenesis , Osteogenesis Imperfecta/drug therapy , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/metabolism , Phenotype
4.
Front Cell Dev Biol ; 10: 963389, 2022.
Article in English | MEDLINE | ID: mdl-36726589

ABSTRACT

Alterations in the balance between skeletogenesis and adipogenesis is a pathogenic feature in multiple skeletal disorders. Clinically, enhanced bone marrow adiposity in bones impairs mobility and increases fracture risk, reducing the quality of life of patients. The molecular mechanism that underlies the balance between skeletogenesis and adipogenesis is not completely understood but alterations in skeletal progenitor cells' differentiation pathway plays a key role. We recently demonstrated that parathyroid hormone (PTH)/PTH-related peptide (PTHrP) control the levels of DEPTOR, an inhibitor of the mechanistic target of rapamycin (mTOR), and that DEPTOR levels are altered in different skeletal diseases. Here, we show that mutations in the PTH receptor-1 (PTH1R) alter the differentiation of skeletal progenitors in two different skeletal genetic disorders and lead to accumulation of fat or cartilage in bones. Mechanistically, DEPTOR controls the subcellular localization of TAZ (transcriptional co-activator with a PDZ-binding domain), a transcriptional regulator that governs skeletal stem cells differentiation into either bone and fat. We show that DEPTOR regulation of TAZ localization is achieved through the control of Dishevelled2 (DVL2) phosphorylation. Depending on nutrient availability, DEPTOR directly interacts with PTH1R to regulate PTH/PTHrP signaling or it forms a complex with TAZ, to prevent its translocation to the nucleus and therefore inhibit its transcriptional activity. Our data point DEPTOR as a key molecule in skeletal progenitor differentiation; its dysregulation under pathologic conditions results in aberrant bone/fat balance.

5.
EBioMedicine ; 62: 103075, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33242826

ABSTRACT

BACKGROUND: Beyond its structural role in the skeleton, the extracellular matrix (ECM), particularly basement membrane proteins, facilitates communication with intracellular signaling pathways and cell to cell interactions to control differentiation, proliferation, migration and survival. Alterations in extracellular proteins cause a number of skeletal disorders, yet the consequences of an abnormal ECM on cellular communication remains less well understood METHODS: Clinical and radiographic examinations defined the phenotype in this unappreciated bent bone skeletal disorder. Exome analysis identified the genetic alteration, confirmed by Sanger sequencing. Quantitative PCR, western blot analyses, immunohistochemistry, luciferase assay for WNT signaling were employed to determine RNA, proteins levels and localization, and dissect out the underlying cell signaling abnormalities.  Migration and wound healing assays examined cell migration properties. FINDINGS: This bent bone dysplasia resulted from biallelic mutations in LAMA5, the gene encoding the alpha-5 laminin basement membrane protein. This finding uncovered a mechanism of disease driven by ECM-cell interactions between alpha-5-containing laminins, and integrin-mediated focal adhesion signaling, particularly in cartilage. Loss of LAMA5 altered ß1 integrin signaling through the non-canonical kinase PYK2 and the skeletal enriched SRC kinase, FYN. Loss of LAMA5 negatively impacted the actin cytoskeleton, vinculin localization, and WNT signaling. INTERPRETATION: This newly described mechanism revealed a LAMA5-ß1 Integrin-PYK2-FYN focal adhesion complex that regulates skeletogenesis, impacted WNT signaling and, when dysregulated, produced a distinct skeletal disorder. FUNDING: Supported by NIH awards R01 AR066124, R01 DE019567, R01 HD070394, and U54HG006493, and Czech Republic grants INTER-ACTION LTAUSA19030, V18-08-00567 and GA19-20123S.


Subject(s)
Alleles , Bone Diseases, Developmental/etiology , Bone Diseases, Developmental/metabolism , Cell Adhesion/genetics , Laminin/genetics , Laminin/metabolism , Mutation , Signal Transduction , Bone Diseases, Developmental/diagnosis , Bone and Bones/abnormalities , Bone and Bones/diagnostic imaging , Chondrocytes/metabolism , DNA Mutational Analysis , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Phenotype , Wnt Signaling Pathway , src-Family Kinases/metabolism
6.
Biomolecules ; 10(2)2020 02 19.
Article in English | MEDLINE | ID: mdl-32092942

ABSTRACT

Skeletal development is a complex process which requires the tight regulation of gene activation and suppression in response to local signaling pathways. Among these pathways, Notch signaling is implicated in governing cell fate determination, proliferation, differentiation and apoptosis of skeletal cells-osteoblasts, osteoclasts, osteocytes and chondrocytes. Moreover, human genetic mutations in Notch components emphasize the critical roles of Notch signaling in skeletal development and homeostasis. In this review, we focus on the physiological roles of Notch signaling in skeletogenesis, postnatal bone and cartilage homeostasis and fracture repair. We also discuss the pathological gain- and loss-of-function of Notch signaling in bone and cartilage, resulting in osteosarcoma and age-related degenerative diseases, such as osteoporosis and osteoarthritis. Understanding the physiological and pathological function of Notch signaling in skeletal tissues using animal models and human genetics will provide new insights into disease pathogenesis and offer novel approaches for the treatment of bone/cartilage diseases.


Subject(s)
Bone Diseases/metabolism , Chondrogenesis , Osteogenesis , Receptors, Notch/metabolism , Signal Transduction , Animals , Bone Diseases/pathology , Bone Diseases/physiopathology , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/physiopathology , Homeostasis , Humans , Stem Cells/metabolism , Stem Cells/pathology
7.
J Bone Miner Res ; 35(6): 1132-1148, 2020 06.
Article in English | MEDLINE | ID: mdl-32053224

ABSTRACT

Osteogenesis imperfecta (OI) is a genetic bone dysplasia characterized by bone deformities and fractures caused by low bone mass and impaired bone quality. OI is a genetically heterogeneous disorder that most commonly arises from dominant mutations in genes encoding type I collagen (COL1A1 and COL1A2). In addition, OI is recessively inherited with the majority of cases resulting from mutations in prolyl-3-hydroxylation complex members, which includes cartilage-associated protein (CRTAP). OI patients are at an increased risk of fracture throughout their lifetimes. However, non-union or delayed healing has been reported in 24% of fractures and 52% of osteotomies. Additionally, refractures typically go unreported, making the frequency of refractures in OI patients unknown. Thus, there is an unmet need to better understand the mechanisms by which OI affects fracture healing. Using an open tibial fracture model, our study demonstrates delayed healing in both Col1a2 G610c/+ and Crtap -/- OI mouse models (dominant and recessive OI, respectively) that is associated with reduced callus size and predicted strength. Callus cartilage distribution and chondrocyte maturation were altered in OI, suggesting accelerated cartilage differentiation. Importantly, we determined that healed fractured tibia in female OI mice are biomechanically weaker when compared with the contralateral unfractured bone, suggesting that abnormal OI fracture healing OI may prime future refracture at the same location. We have previously shown upregulated TGF-ß signaling in OI and we confirm this in the context of fracture healing. Interestingly, treatment of Crtap -/- mice with the anti-TGF-ß antibody 1D11 resulted in further reduced callus size and predicted strength, highlighting the importance of investigating dose response in treatment strategies. These data provide valuable insight into the effect of the extracellular matrix (ECM) on fracture healing, a poorly understood mechanism, and support the need for prevention of primary fractures to decrease incidence of refracture and deformity in OI patients. © 2020 American Society for Bone and Mineral Research.


Subject(s)
Osteogenesis Imperfecta , Animals , Collagen , Collagen Type I/genetics , Extracellular Matrix Proteins , Female , Fracture Healing , Humans , Mice , Molecular Chaperones , Osteogenesis Imperfecta/genetics
8.
Proc Natl Acad Sci U S A ; 116(10): 4316-4325, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30782830

ABSTRACT

Vertebrate primary cilium is a Hedgehog signaling center but the extent of its involvement in other signaling systems is less well understood. This report delineates a mechanism by which fibroblast growth factor (FGF) controls primary cilia. Employing proteomic approaches to characterize proteins associated with the FGF-receptor, FGFR3, we identified the serine/threonine kinase intestinal cell kinase (ICK) as an FGFR interactor. ICK is involved in ciliogenesis and participates in control of ciliary length. FGF signaling partially abolished ICK's kinase activity, through FGFR-mediated ICK phosphorylation at conserved residue Tyr15, which interfered with optimal ATP binding. Activation of the FGF signaling pathway affected both primary cilia length and function in a manner consistent with cilia effects caused by inhibition of ICK activity. Moreover, knockdown and knockout of ICK rescued the FGF-mediated effect on cilia. We provide conclusive evidence that FGF signaling controls cilia via interaction with ICK.


Subject(s)
Cilia/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , CRISPR-Cas Systems , Fibroblast Growth Factors/metabolism , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Knockout , Models, Animal , Molecular Docking Simulation , NIH 3T3 Cells , Phosphorylation , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Proteomics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction
9.
Sci Signal ; 11(548)2018 09 18.
Article in English | MEDLINE | ID: mdl-30228226

ABSTRACT

Sustained activation of extracellular signal-regulated kinase (ERK) drives pathologies caused by mutations in fibroblast growth factor receptors (FGFRs). We previously identified the inositol phosphatase SHIP2 (also known as INPPL1) as an FGFR-interacting protein and a target of the tyrosine kinase activities of FGFR1, FGFR3, and FGFR4. We report that loss of SHIP2 converted FGF-mediated sustained ERK activation into a transient signal and rescued cell phenotypes triggered by pathologic FGFR-ERK signaling. Mutant forms of SHIP2 lacking phosphoinositide phosphatase activity still associated with FGFRs and did not prevent FGF-induced sustained ERK activation, demonstrating that the adaptor rather than the catalytic activity of SHIP2 was required. SHIP2 recruited Src family kinases to the FGFRs, which promoted FGFR-mediated phosphorylation and assembly of protein complexes that relayed signaling to ERK. SHIP2 interacted with FGFRs, was phosphorylated by active FGFRs, and promoted FGFR-ERK signaling at the level of phosphorylation of the adaptor FRS2 and recruitment of the tyrosine phosphatase PTPN11. Thus, SHIP2 is an essential component of canonical FGF-FGFR signal transduction and a potential therapeutic target in FGFR-related disorders.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptors, Fibroblast Growth Factor/genetics , src-Family Kinases/genetics
10.
Sci Rep ; 7: 41803, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28205584

ABSTRACT

Spondylocarpotarsal synostosis (SCT) is a skeletal disorder characterized by progressive vertebral, carpal and tarsal fusions, and mild short stature. The majority of affected individuals have an autosomal recessive form of SCT and are homozygous or compound heterozygous for nonsense mutations in the gene that encodes the cytoskeletal protein filamin B (FLNB), but a subset do not have FLNB mutations. Exome sequence analysis of three SCT patients negative for FLNB mutations identified an autosomal dominant form of the disease due to heterozygosity for missense or nonsense mutations in MYH3, which encodes embryonic myosin. Cells transfected with the MYH3 missense mutations had reduced TGFß signaling, revealing a regulatory role for embryonic myosin in the TGFß signaling pathway. In wild-type mice, there was persistent postnatal expression of embryonic myosin in the small muscles joining the neural arches of the spine suggesting that loss of myosin function in these muscles contribute to the disease. Our findings demonstrate that dominant mutations in MYH3 underlie autosomal dominant SCT, identify a postnatal role for embryonic myosin and suggest that altered regulation of signal transduction in the muscles within the spine may lead to the development of vertebral fusions.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Cytoskeletal Proteins/genetics , Genes, Dominant , Lumbar Vertebrae/abnormalities , Musculoskeletal Diseases/genetics , Musculoskeletal Diseases/metabolism , Mutation , Myosins/genetics , Myosins/metabolism , Scoliosis/congenital , Signal Transduction , Synostosis/genetics , Synostosis/metabolism , Thoracic Vertebrae/abnormalities , Transforming Growth Factor beta/metabolism , Abnormalities, Multiple/diagnosis , Alleles , Bone Morphogenetic Proteins/metabolism , Female , Genotype , Humans , Lumbar Vertebrae/metabolism , Male , Musculoskeletal Diseases/diagnosis , Phenotype , Radiography , Scoliosis/diagnosis , Scoliosis/genetics , Scoliosis/metabolism , Synostosis/diagnosis , Thoracic Vertebrae/metabolism , Exome Sequencing
11.
PLoS Genet ; 12(3): e1005936, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27019229

ABSTRACT

Spondylocarpotarsal synostosis (SCT) is an autosomal recessive disorder characterized by progressive vertebral fusions and caused by loss of function mutations in Filamin B (FLNB). FLNB acts as a signaling scaffold by linking the actin cytoskleteon to signal transduction systems, yet the disease mechanisms for SCT remain unclear. Employing a Flnb knockout mouse, we found morphologic and molecular evidence that the intervertebral discs (IVDs) of Flnb-/-mice undergo rapid and progressive degeneration during postnatal development as a result of abnormal cell fate changes in the IVD, particularly the annulus fibrosus (AF). In Flnb-/-mice, the AF cells lose their typical fibroblast-like characteristics and acquire the molecular and phenotypic signature of hypertrophic chondrocytes. This change is characterized by hallmarks of endochondral-like ossification including alterations in collagen matrix, expression of Collagen X, increased apoptosis, and inappropriate ossification of the disc tissue. We show that conversion of the AF cells into chondrocytes is coincident with upregulated TGFß signaling via Smad2/3 and BMP induced p38 signaling as well as sustained activation of canonical and noncanonical target genes p21 and Ctgf. These findings indicate that FLNB is involved in attenuation of TGFß/BMP signaling and influences AF cell fate. Furthermore, we demonstrate that the IVD disruptions in Flnb-/-mice resemble aging degenerative discs and reveal new insights into the molecular causes of vertebral fusions and disc degeneration.


Subject(s)
Abnormalities, Multiple/genetics , Filamins/genetics , Intervertebral Disc Degeneration/genetics , Lumbar Vertebrae/abnormalities , Musculoskeletal Diseases/genetics , Scoliosis/congenital , Synostosis/genetics , Thoracic Vertebrae/abnormalities , Transforming Growth Factor beta/genetics , Abnormalities, Multiple/pathology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Chondrocytes/metabolism , Chondrocytes/pathology , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Disease Models, Animal , Growth Plate/growth & development , Growth Plate/pathology , Humans , Intervertebral Disc Degeneration/pathology , Lumbar Vertebrae/pathology , Mice , Mice, Knockout , Musculoskeletal Diseases/pathology , Scoliosis/genetics , Scoliosis/pathology , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , Spine/growth & development , Spine/pathology , Synostosis/pathology , Thoracic Vertebrae/pathology
12.
Genome Res ; 24(7): 1224-35, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24985916

ABSTRACT

Annotation of regulatory elements and identification of the transcription-related factors (TRFs) targeting these elements are key steps in understanding how cells interpret their genetic blueprint and their environment during development, and how that process goes awry in the case of disease. One goal of the modENCODE (model organism ENCyclopedia of DNA Elements) Project is to survey a diverse sampling of TRFs, both DNA-binding and non-DNA-binding factors, to provide a framework for the subsequent study of the mechanisms by which transcriptional regulators target the genome. Here we provide an updated map of the Drosophila melanogaster regulatory genome based on the location of 84 TRFs at various stages of development. This regulatory map reveals a variety of genomic targeting patterns, including factors with strong preferences toward proximal promoter binding, factors that target intergenic and intronic DNA, and factors with distinct chromatin state preferences. The data also highlight the stringency of the Polycomb regulatory network, and show association of the Trithorax-like (Trl) protein with hotspots of DNA binding throughout development. Furthermore, the data identify more than 5800 instances in which TRFs target DNA regions with demonstrated enhancer activity. Regions of high TRF co-occupancy are more likely to be associated with open enhancers used across cell types, while lower TRF occupancy regions are associated with complex enhancers that are also regulated at the epigenetic level. Together these data serve as a resource for the research community in the continued effort to dissect transcriptional regulatory mechanisms directing Drosophila development.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , Genome, Insect , Transcription Factors , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Chromatin/genetics , Chromatin/metabolism , Cluster Analysis , Computational Biology/methods , Enhancer Elements, Genetic , Gene Expression Profiling , Genomics/methods , Nucleotide Motifs , Protein Binding , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
13.
Nat Methods ; 9(6): 609-14, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22522655

ABSTRACT

We evaluated how variations in sequencing depth and other parameters influence interpretation of chromatin immunoprecipitation-sequencing (ChIP-seq) experiments. Using Drosophila melanogaster S2 cells, we generated ChIP-seq data sets for a site-specific transcription factor (Suppressor of Hairy-wing) and a histone modification (H3K36me3). We detected a chromatin-state bias: open chromatin regions yielded higher coverage, which led to false positives if not corrected. This bias had a greater effect on detection specificity than any base-composition bias. Paired-end sequencing revealed that single-end data underestimated ChIP-library complexity at high coverage. Removal of reads originating at the same base reduced false-positives but had little effect on detection sensitivity. Even at mappable-genome coverage depth of ∼1 read per base pair, ∼1% of the narrow peaks detected on a tiling array were missed by ChIP-seq. Evaluation of widely used ChIP-seq analysis tools suggests that adjustments or algorithm improvements are required to handle data sets with deep coverage.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/chemistry , Algorithms , Animals , Chromatin Immunoprecipitation/standards , Drosophila Proteins/genetics , Drosophila melanogaster , False Positive Reactions , Gene Library , High-Throughput Nucleotide Sequencing , Histone-Lysine N-Methyltransferase/genetics , Oligonucleotide Array Sequence Analysis , Repressor Proteins/genetics , Sensitivity and Specificity
14.
BMC Genet ; 12: 54, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21639918

ABSTRACT

BACKGROUND: Genetic interactions within hybrids influence their overall fitness. Understanding the details of these interactions can improve our understanding of speciation. One experimental approach is to investigate deviations from Mendelian expectations (segregation distortion) in the inheritance of mapped genetic markers. In this study, we used the copepod Tigriopus californicus, a species which exhibits high genetic divergence between populations and a general pattern of reduced fitness in F2 interpopulation hybrids. Previous studies have implicated both nuclear-cytoplasmic and nuclear-nuclear interactions in causing this fitness reduction. We identified and mapped population-diagnostic single nucleotide polymorphisms (SNPs) and used these to examine segregation distortion across the genome within F2 hybrids. RESULTS: We generated a linkage map which included 45 newly elucidated SNPs and 8 population-diagnostic microsatellites used in previous studies. The map, the first available for the Copepoda, was estimated to cover 75% of the genome and included markers on all 12 T. californicus chromosomes. We observed little segregation distortion in newly hatched F2 hybrid larvae (fewer than 10% of markers at p < 0.05), but strikingly higher distortion in F2 hybrid adult males (45% of markers at p < 0.05). Hence, segregation distortion was primarily caused by selection against particular genetic combinations which acted between hatching and maturity. Distorted markers were not distributed randomly across the genome but clustered on particular chromosomes. In contrast to other studies in this species we found little evidence for cytonuclear coadaptation. Instead, different linkage groups exhibited markedly different patterns of distortion, which appear to have been influenced by nuclear-nuclear epistatic interactions and may also reflect genetic load carried within the parental lines. CONCLUSION: Adult male F2 hybrids between two populations of T. californius exhibit dramatic segregation distortion across the genome. Distorted loci are clustered within specific linkage groups, and the direction of distortion differs between chromosomes. This segregation distortion is due to selection acting between hatching and adulthood.


Subject(s)
Copepoda/genetics , Hybridization, Genetic , Inheritance Patterns , Polymorphism, Single Nucleotide , Animals , Cell Nucleus/genetics , Chromosome Mapping/methods , Chromosomes/genetics , Crosses, Genetic , Female , Gene Library , Genetic Loci , Genome , Genotype , Larva/genetics , Male , Microsatellite Repeats , Mitochondria/genetics , Selection, Genetic , Sexual Behavior, Animal
15.
Nature ; 471(7339): 527-31, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21430782

ABSTRACT

Systematic annotation of gene regulatory elements is a major challenge in genome science. Direct mapping of chromatin modification marks and transcriptional factor binding sites genome-wide has successfully identified specific subtypes of regulatory elements. In Drosophila several pioneering studies have provided genome-wide identification of Polycomb response elements, chromatin states, transcription factor binding sites, RNA polymerase II regulation and insulator elements; however, comprehensive annotation of the regulatory genome remains a significant challenge. Here we describe results from the modENCODE cis-regulatory annotation project. We produced a map of the Drosophila melanogaster regulatory genome on the basis of more than 300 chromatin immunoprecipitation data sets for eight chromatin features, five histone deacetylases and thirty-eight site-specific transcription factors at different stages of development. Using these data we inferred more than 20,000 candidate regulatory elements and validated a subset of predictions for promoters, enhancers and insulators in vivo. We identified also nearly 2,000 genomic regions of dense transcription factor binding associated with chromatin activity and accessibility. We discovered hundreds of new transcription factor co-binding relationships and defined a transcription factor network with over 800 potential regulatory relationships.


Subject(s)
Drosophila melanogaster/genetics , Genome, Insect/genetics , Molecular Sequence Annotation , Regulatory Sequences, Nucleic Acid/genetics , Animals , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Enhancer Elements, Genetic/genetics , Histone Deacetylases/metabolism , Insulator Elements/genetics , Promoter Regions, Genetic/genetics , Reproducibility of Results , Silencer Elements, Transcriptional/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...