Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Eur Rev Med Pharmacol Sci ; 28(8): 3216-3226, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38708480

ABSTRACT

OBJECTIVE: The study aims to provide guidance on the identification of multiple-digit malformations as potential biomarkers and therapeutic targets. MATERIALS AND METHODS: Single-cell RNA sequencing (scRNA-seq) data of four multiple-finger malformation samples were downloaded from the GEO public database. Fibroblasts and keratinocytes were divided into cellular subpopulations and the transcription factors of different subpopulations were analyzed. The regulatory network of transcription factors and their target genes were constructed to analyze the functionality of regulons. RESULTS: Examination of the transcriptional profile data from 11,806 single cells uncovered significant associations between regulons and cell function in polydactyly. Specifically, the analysis highlighted the involvement of HOX family members and GLI2 transcription factors, including HOXD13, MSX2, LHX2, EMX2, LEF1, CREB3L2, and LHX2, in the polydactyly process within fibroblast cells. Furthermore, it sheds light on the roles of HES2 and GLIS1 in the formation and development of keratinocytes. CONCLUSIONS: Significant presence of transcription factors, especially HOXD13, MSX2, and LHX2, may be strongly related to the development of polydactyly.


Subject(s)
Polydactyly , Single-Cell Analysis , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Humans , Polydactyly/genetics , Polydactyly/pathology , Polydactyly/metabolism , Gene Expression Profiling , Fibroblasts/metabolism , Keratinocytes/metabolism , Transcriptome , Single-Cell Gene Expression Analysis
2.
Arthritis Res Ther ; 26(1): 66, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38468277

ABSTRACT

BACKGROUND: CRISPR-Cas9-based genome engineering represents a powerful therapeutic tool for cartilage tissue engineering and for understanding molecular pathways driving cartilage diseases. However, primary chondrocytes are difficult to transfect and rapidly dedifferentiate during monolayer (2D) cell culture, making the lengthy expansion of a single-cell-derived edited clonal population not feasible. For this reason, functional genetics studies focused on cartilage and rheumatic diseases have long been carried out in cellular models that poorly recapitulate the native molecular properties of human cartilaginous tissue (e.g., cell lines, induced pluripotent stem cells). Here, we set out to develop a non-viral CRISPR-Cas9, bulk-gene editing method suitable for chondrocyte populations from different cartilaginous sources. METHODS: We screened electroporation and lipid nanoparticles for ribonucleoprotein (RNP) delivery in primary polydactyly chondrocytes, and optimized RNP reagents assembly. We knocked out RELA (also known as p65), a subunit of the nuclear factor kappa B (NF-κB), in polydactyly chondrocytes and further characterized knockout (KO) cells with RT-qPCR and Western Blot. We tested RELA KO in chondrocytes from diverse cartilaginous sources and characterized their phenotype with RT-qPCR. We examined the chondrogenic potential of wild-type (WT) and KO cell pellets in presence and absence of interleukin-1ß (IL-1ß). RESULTS: We established electroporation as the optimal transfection technique for chondrocytes enhancing transfection and editing efficiency, while preserving high cell viability. We knocked out RELA with an unprecedented efficiency of ~90%, confirming lower inflammatory pathways activation upon IL-1ß stimulation compared to unedited cells. Our protocol could be easily transferred to primary human chondrocytes harvested from osteoarthritis (OA) patients, human FE002 chondroprogenitor cells, bovine chondrocytes, and a human chondrocyte cell line, achieving comparable mean RELA KO editing levels using the same protocol. All KO pellets from primary human chondrocytes retained chondrogenic ability equivalent to WT cells, and additionally displayed enhanced matrix retention under inflamed conditions. CONCLUSIONS: We showcased the applicability of our bulk gene editing method to develop effective autologous and allogeneic off-the-shelf gene therapies strategies and to enable functional genetics studies in human chondrocytes to unravel molecular mechanisms of cartilage diseases.


Subject(s)
Cartilage Diseases , Polydactyly , Humans , Animals , Cattle , Chondrocytes/metabolism , Gene Editing/methods , CRISPR-Cas Systems/genetics , Interleukin-1beta/metabolism , Cartilage Diseases/metabolism , Polydactyly/metabolism
3.
Cell Transplant ; 33: 9636897231221878, 2024.
Article in English | MEDLINE | ID: mdl-38164917

ABSTRACT

This study compared the proliferation and differentiation potential of bone marrow-derived mesenchymal stem cells (BMSCs) derived from infants with polydactyly and adults with basal joint arthritis. The proliferation rate of adult and infant BMSCs was determined by the cell number changes and doubling times. The γH2AX immunofluorescence staining, age-related gene expression, senescence-associated ß-galactosidase (SA-ß-gal) staining were analyzed to determine the senescence state of adult and infant BMSCs. The expression levels of superoxide dismutases (SODs) and genes associated with various types of differentiation were measured using Real-Time Quantitative Polymerase Chain Reaction (RT-qPCR). Differentiation levels were evaluated through histochemical and immunohistochemical staining. The results showed that infant BMSCs had a significantly higher increase in cell numbers and faster doubling times compared with adult BMSCs. Infant BMSCs at late stages exhibited reduced γH2AX expression and SA-ß-gal staining, indicating lower levels of senescence. The expression levels of senescence-related genes (p16, p21, and p53) in infant BMSCs were also lower than in adult BMSCs. In addition, infant BMSCs demonstrated higher antioxidative ability with elevated expression of SOD1, SOD2, and SOD3 compared with adult BMSCs. In terms of differentiation potential, infant BMSCs outperformed adult BMSCs in chondrogenesis, as indicated by higher expression levels of chondrogenic genes (SOX9, COL2, and COL10) and positive immunohistochemical staining. Moreover, differentiated cells derived from infant BMSCs exhibited significantly higher expression levels of osteogenic, tenogenic, hepatogenic, and neurogenic genes compared with those derived from adult BMSCs. Histochemical and immunofluorescence staining confirmed these findings. However, adult BMSCs showed lower adipogenic differentiation potential compared with infant BMSCs. Overall, infant BMSCs demonstrated superior characteristics, including higher proliferation rates, enhanced antioxidative activity, and greater differentiation potential into various lineages. They also exhibited reduced cellular senescence. These findings, within the context of cellular differentiation, suggest potential implications for the use of allogeneic BMSC transplantation, emphasizing the need for further in vivo investigation.


Subject(s)
Arthritis , Mesenchymal Stem Cells , Polydactyly , Adult , Child , Humans , Bone Marrow , Cell Proliferation , Cell Differentiation , Osteogenesis/genetics , Cells, Cultured , Bone Marrow Cells , Arthritis/metabolism , Polydactyly/metabolism
4.
Dev Cell ; 57(19): 2273-2289.e11, 2022 10 10.
Article in English | MEDLINE | ID: mdl-36220081

ABSTRACT

Digit determination in limb buds is driven by a posteriorizing Sonic hedgehog (Shh) protein gradient; however, the mechanism regulating this is unclear. Here, we propose a diffusion-and-trapping hypothesis for Shh gradient formation based on data from the preaxial polydactyly phenotype of KIF3B motor hypomorphic mice. In the limb buds of these mice, a distal-to-proximal gradient of fibroblast growth factor (FGF) and phosphatidylinositol 3-kinase (PI3K) signaling and a posterior-to-anterior gradient of Shh were disorganized. This phenotype was reproduced by transplanting FGF8b-soaked beads. At the subcellular level, KIF3B transported the phosphatase and tensin homolog (PTEN)-like phosphatase Talpid3 to terminate PI3K signaling. High and low PI3K signaling strengths differentially sorted endocytosed Shh toward exosome-like particles and cytonemal punctata, respectively. These results indicate that the Shh-containing particles undergo either the diffusional movement in the periphery or cytonemal trapping in the center and form a spatial gradient along the periphery of developing limb buds.


Subject(s)
Hedgehog Proteins , Polydactyly , Animals , Extremities , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Kinesins , Limb Buds/metabolism , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphoric Monoester Hydrolases/genetics , Polydactyly/genetics , Polydactyly/metabolism , Tensins/genetics , Tensins/metabolism
5.
Int J Mol Sci ; 22(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34502207

ABSTRACT

The complexity of skeletal pathologies makes use of in vivo models essential to elucidate the pathogenesis of the diseases; nevertheless, chondrocyte and osteoblast cell lines provide relevant information on the underlying disease mechanisms. Due to the limitations of primary chondrocytes, immortalized cells represent a unique tool to overcome this problem since they grow very easily for several passages. However, in the immortalization procedure the cells might lose the original phenotype; thus, these cell lines should be deeply characterized before their use. We immortalized primary chondrocytes from a Cant1 knock-out mouse, an animal model of Desbuquois dysplasia type 1, with a plasmid expressing the SV40 large and small T antigen. This cell line, based on morphological and biochemical parameters, showed preservation of the chondrocyte phenotype. In addition reduced proteoglycan synthesis and oversulfation of glycosaminoglycan chains were demonstrated, as already observed in primary chondrocytes from the Cant1 knock-out mouse. In conclusion, immortalized Cant1 knock-out chondrocytes maintained the disease phenotype observed in primary cells validating the in vitro model and providing an additional tool to further study the proteoglycan biosynthesis defect. The same approach might be extended to other cartilage disorders.


Subject(s)
Acid Anhydride Hydrolases/physiology , Chondrocytes/pathology , Craniofacial Abnormalities/pathology , Dwarfism/pathology , Glycosaminoglycans/metabolism , Joint Instability/pathology , Ossification, Heterotopic/pathology , Phenotype , Polydactyly/pathology , Animals , Cell Line, Transformed , Chondrocytes/metabolism , Craniofacial Abnormalities/etiology , Craniofacial Abnormalities/metabolism , Dwarfism/etiology , Dwarfism/metabolism , Joint Instability/etiology , Joint Instability/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ossification, Heterotopic/etiology , Ossification, Heterotopic/metabolism , Polydactyly/etiology , Polydactyly/metabolism
6.
PLoS One ; 15(8): e0237985, 2020.
Article in English | MEDLINE | ID: mdl-32822395

ABSTRACT

Allogeneic cultured epidermis (allo-CE) is a cultured keratinocyte sheet manufactured from donor cells and promotes wound healing when used in deep dermal burns, donor sites, and chronic ulcers and serves as a wound dressing. Allo-CE is usually cryopreserved to be ready to use. However, the cryopreservation procedure will damage the cell viability, and the influence of Allo-CE, according to its viability or wound healing process, has not been evaluated sufficiently. In this study, we aimed to prove the influence of keratinocyte viability contained in allo-CEs on wound healing. We prepared CEs with Green's method using keratinocytes obtained from a polydactyly patient and then prepared four kinds of CEs with different cell viabilities [fresh, cryopreserved, frozen, and FT (freeze and thaw)]. The cell viabilities of fresh, cryopreserved, frozen, and FT CEs were 95.7%, 59.9%, 16.7%, and 0.0%, respectively. The four CEs had homogeneous characteristics, except for small gaps found in the FT sheet by transmission electron microscopy observation. The four CEs were applied on the full-thickness skin defect of diabetic mice (BKS.Cg-Dock 7m +/+ Leprdb/Jcl), and the wound area and neoepithelium length were evaluated on days 4, 7, and 14. As a result, FT CEs without viable cells similarly promoted epithelialization on days 4 and 7 (p<0.05) and accelerated wound closure on day 7 (p<0.01) as fresh CEs compared with the control group. In conclusion, the promoting effect of allo-CE on wound healing does not depend on cell viability. Lyophilized CEs may be a suitable wound dressing with a long storage period at room temperature.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Keratinocytes/transplantation , Wound Healing , Animals , Cell Survival , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Humans , Infant , Keratinocytes/cytology , Keratinocytes/metabolism , Male , Mice , Polydactyly/metabolism , Polydactyly/pathology , Re-Epithelialization
7.
Cell ; 180(6): 1262-1271.e15, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32169219

ABSTRACT

Establishing causal links between non-coding variants and human phenotypes is an increasing challenge. Here, we introduce a high-throughput mouse reporter assay for assessing the pathogenic potential of human enhancer variants in vivo and examine nearly a thousand variants in an enhancer repeatedly linked to polydactyly. We show that 71% of all rare non-coding variants previously proposed as causal lead to reporter gene expression in a pattern consistent with their pathogenic role. Variants observed to alter enhancer activity were further confirmed to cause polydactyly in knockin mice. We also used combinatorial and single-nucleotide mutagenesis to evaluate the in vivo impact of mutations affecting all positions of the enhancer and identified additional functional substitutions, including potentially pathogenic variants hitherto not observed in humans. Our results uncover the functional consequences of hundreds of mutations in a phenotype-associated enhancer and establish a widely applicable strategy for systematic in vivo evaluation of human enhancer variants.


Subject(s)
Enhancer Elements, Genetic/genetics , High-Throughput Screening Assays/methods , Polydactyly/genetics , Animals , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Developmental/genetics , Gene Knock-In Techniques/methods , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Mice , Mutation , Phenotype , Polydactyly/metabolism , RNA, Untranslated/genetics
8.
Genet Med ; 22(1): 189-198, 2020 01.
Article in English | MEDLINE | ID: mdl-31395945

ABSTRACT

PURPOSE: Preaxial polydactyly (PPD) is a common congenital hand malformation classified into four subtypes (PPD I-IV). Variants in the zone of polarizing activity regulatory sequence (ZRS) within intron 5 of the LMBR1 gene are linked to most PPD types. However, the genes responsible for PPD I and the underlying mechanisms are unknown. METHODS: A rare large four-generation family with isolated PPD I was subjected to genome-wide genotyping and sequence analysis. In vitro and in vivo functional studies were performed in Caco-2 cells, 293T cells, and a knockin transgenic mouse model. RESULTS: A novel g.101779T>A (reference sequence: NG_009240.2; position 446 of the ZRS) variant segregates with all PPD I-affected individuals. The knockin mouse with this ZRS variant exhibited PPD I phenotype accompanying ectopic and excess expression of Shh. We confirmed that HnRNP K can bind the ZRS and SHH promoters. The ZRS mutant enhanced the binding affinity for HnRNP K and upregulated SHH expression. CONCLUSION: Our results identify the first PPD I disease-causing variant. The variant leading to PPD I may be associated with enhancing SHH expression mediated by HnRNP K. This study adds to the ZRS-associated syndromes classification system for PPD and clarifies the underlying molecular mechanisms.


Subject(s)
Hedgehog Proteins/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Limb Buds/growth & development , Membrane Proteins/genetics , Polydactyly/genetics , Polymorphism, Single Nucleotide , Thumb/abnormalities , Up-Regulation , Animals , Caco-2 Cells , Disease Models, Animal , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Introns , Limb Buds/metabolism , Limb Buds/pathology , Male , Mice , Mice, Transgenic , Pedigree , Polydactyly/metabolism
9.
J Cutan Pathol ; 46(7): 490-497, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30893489

ABSTRACT

BACKGROUNDS: We previously demonstrated the presence of onychodermis below nail matrix and nail bed. Because nail matrix is a producer of nail plate, we hypothesized that onychodermis below nail matrix could be the nail counterpart of follicular dermal papilla. In this study, we sought to further characterize histologic, histochemical, and immunohistochemical features of nail matrix onychodermis. METHODS AND RESULTS: Hematoxylin and eosin slides of 10 polydactyly nail units and 10 nail matrix biopsies from children and adults were reviewed. In polydactyly nail units, the onychodermis beneath nail matrix was characterized by onychofibroblasts showing abundant cytoplasm, and this area was slightly separated from the undersurface of the nail matrix. Nail matrix biopsy specimens also showed similar histology in the nail matrix onychodermis. Alcian blue stain demonstrated mucin deposition in onychofibroblasts within the nail matrix onychodermis. Immunohistochemically, elastin was rarely expressed in the nail matrix onychodermis while it was strongly expressed in the dermis of other areas of polydactyly nail units. Elastin was not expressed in follicular dermal papilla of terminal hair follicles of the scalp. CONCLUSION: Our findings demonstrate the presence and localization of nail matrix onychodermis (onychomatricodermis). Our study also demonstrates similar elastin expression patterns in the onychomatricodermis and follicular dermal papilla.


Subject(s)
Dermis , Hair Follicle , Nails , Polydactyly , Dermis/metabolism , Dermis/pathology , Female , Hair Follicle/metabolism , Hair Follicle/pathology , Humans , Immunohistochemistry , Male , Nails/metabolism , Nails/pathology , Polydactyly/metabolism , Polydactyly/pathology
10.
J Bone Miner Res ; 34(2): 375-386, 2019 02.
Article in English | MEDLINE | ID: mdl-30395363

ABSTRACT

Polydactyly is a common congenital anomaly of the hand and foot. Postaxial polydactyly (PAP) is characterized by one or more posterior or postaxial digits. In a Pakistani family with autosomal recessive nonsyndromic postaxial polydactyly type A (PAPA), we performed genomewide genotyping, linkage analysis, and exome and Sanger sequencing. Exome sequencing revealed a homozygous nonsense variant (c.478C>T, p.[Arg160*]) in the FAM92A gene within the mapped region on 8q21.13-q24.12 that segregated with the PAPA phenotype. We found that FAM92A is expressed in the developing mouse limb and E11.5 limb bud including the progress zone and the apical ectodermal ridge, where it strongly localizes at the cilia level, suggesting an important role in limb patterning. The identified variant leads to a loss of the FAM92A/Chibby1 complex that is crucial for ciliogenesis and impairs the recruitment and the colocalization of FAM92A with Chibby1 at the base of the cilia. In addition, we show that Fam92a-/- homozygous mice also exhibit an abnormal digit morphology, including metatarsal osteomas and polysyndactyly, in addition to distinct abnormalities on the deltoid tuberosity of their humeri. In conclusion, we present a new nonsyndromic PAPA ciliopathy due to a loss-of-function variant in FAM92A. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Ciliopathies , Codon, Nonsense , Exome , Fingers/abnormalities , Homozygote , Polydactyly , Proteins , Toes/abnormalities , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Female , Fingers/pathology , Humans , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polydactyly/genetics , Polydactyly/metabolism , Polydactyly/pathology , Proteins/genetics , Proteins/metabolism , Toes/pathology , Exome Sequencing
11.
Matrix Biol ; 81: 70-90, 2019 08.
Article in English | MEDLINE | ID: mdl-30439444

ABSTRACT

Desbuquois dysplasia type 1 (DBQD1) is a chondrodysplasia caused by mutations in CANT1 gene encoding an ER/Golgi calcium activated nucleotidase 1 that hydrolyses UDP. Here, using Cant1 knock-in and knock-out mice recapitulating DBQD1 phenotype, we report that CANT1 plays a crucial role in cartilage proteoglycan synthesis and in endochondral ossification. Specifically, the glycosaminoglycan synthesis was decreased in chondrocytes from Cant1 knock-out mice and their hydrodynamic size was reduced, whilst the sulfation was increased and the overall proteoglycan secretion was delayed. Interestingly, knock-out chondrocytes had dilated ER cisternae suggesting delayed protein secretion and cellular stress; however, no canonical ER stress response was detected using microarray analysis, Xbp1 splicing and protein levels of BiP and ATF4. The observed proteoglycan defects caused deregulated chondrocyte proliferation and maturation in the growth plate resulting in the reduced skeletal growth. In conclusion, the pathogenic mechanism of DBQD1 comprises deregulated chondrocyte performance due to defective intracellular proteoglycan synthesis and altered proteoglycan properties in the extracellular matrix.


Subject(s)
Acid Anhydride Hydrolases/genetics , Cartilage/metabolism , Craniofacial Abnormalities/genetics , Dwarfism/genetics , Glycosaminoglycans/biosynthesis , Joint Instability/genetics , Nucleotidases/genetics , Ossification, Heterotopic/genetics , Osteogenesis , Polydactyly/genetics , Animals , Cartilage/cytology , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Craniofacial Abnormalities/metabolism , Disease Models, Animal , Dwarfism/metabolism , Endoplasmic Reticulum/metabolism , Gene Knock-In Techniques , Gene Knockdown Techniques , Humans , Joint Instability/metabolism , Mice , Ossification, Heterotopic/metabolism , Polydactyly/metabolism
12.
Hum Mol Genet ; 26(23): 4556-4571, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973407

ABSTRACT

GLI1, GLI2 and GLI3 form a family of transcription factors which regulate development by mediating the action of Hedgehog (Hh) morphogens. Accordingly, inactivating variants in GLI2 and GLI3 are found in several developmental disorders. In contrast, loss-of-function mutations in GLI1 have remained elusive, maintaining enigmatic the role of this gene in the human embryo. We describe eight patients from three independent families having biallelic truncating variants in GLI1 and developmental defects overlapping with Ellis-van Creveld syndrome (EvC), a disease caused by diminished Hh signaling. Two families had mutations in the last exon of the gene and a third family was identified with an N-terminal stop gain variant predicted to be degraded by the NMD-pathway. Analysis of fibroblasts from one of the patients with homozygous C-terminal truncation of GLI1 demonstrated that the corresponding mutant GLI1 protein is fabricated by patient cells and becomes upregulated in response to Hh signaling. However, the transcriptional activity of the truncated GLI1 factor was found to be severely impaired by cell culture and in vivo assays, indicating that the balance between GLI repressors and activators is altered in affected subjects. Consistent with this, reduced expression of the GLI target PTCH1 was observed in patient fibroblasts after chemical induction of the Hh pathway. We conclude that GLI1 inactivation is associated with a phenotypic spectrum extending from isolated postaxial polydactyly to an EvC-like condition.


Subject(s)
Ellis-Van Creveld Syndrome/genetics , Zinc Finger Protein GLI1/genetics , Child , Ellis-Van Creveld Syndrome/metabolism , Ellis-Van Creveld Syndrome/pathology , Exons , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Developmental , Gene Silencing , Hedgehog Proteins/metabolism , Humans , Infant , Infant, Newborn , Male , Pedigree , Phenotype , Polydactyly/genetics , Polydactyly/metabolism , Primary Cell Culture , Signal Transduction , Trans-Activators/genetics , Transcription, Genetic , Zinc Finger Protein GLI1/metabolism
13.
PLoS Genet ; 13(7): e1006936, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28753627

ABSTRACT

Mutations in BBS6 cause two clinically distinct syndromes, Bardet-Biedl syndrome (BBS), a syndrome caused by defects in cilia transport and function, as well as McKusick-Kaufman syndrome, a genetic disorder characterized by congenital heart defects. Congenital heart defects are rare in BBS, and McKusick-Kaufman syndrome patients do not develop retinitis pigmentosa. Therefore, the McKusick-Kaufman syndrome allele may highlight cellular functions of BBS6 distinct from the presently understood functions in the cilia. In support, we find that the McKusick-Kaufman syndrome disease-associated allele, BBS6H84Y; A242S, maintains cilia function. We demonstrate that BBS6 is actively transported between the cytoplasm and nucleus, and that BBS6H84Y; A242S, is defective in this transport. We developed a transgenic zebrafish with inducible bbs6 to identify novel binding partners of BBS6, and we find interaction with the SWI/SNF chromatin remodeling protein Smarcc1a (SMARCC1 in humans). We demonstrate that through this interaction, BBS6 modulates the sub-cellular localization of SMARCC1 and find, by transcriptional profiling, similar transcriptional changes following smarcc1a and bbs6 manipulation. Our work identifies a new function for BBS6 in nuclear-cytoplasmic transport, and provides insight into the disease mechanism underlying the congenital heart defects in McKusick-Kaufman syndrome patients.


Subject(s)
Abnormalities, Multiple/genetics , Bardet-Biedl Syndrome/genetics , Group II Chaperonins/genetics , Heart Defects, Congenital/genetics , Hydrocolpos/genetics , Polydactyly/genetics , Transcription Factors/genetics , Uterine Diseases/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Active Transport, Cell Nucleus/genetics , Animals , Animals, Genetically Modified/genetics , Bardet-Biedl Syndrome/metabolism , Bardet-Biedl Syndrome/pathology , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Cilia/metabolism , Cilia/pathology , Cytoplasm/metabolism , Disease Models, Animal , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Humans , Hydrocolpos/metabolism , Hydrocolpos/pathology , Mice , Mutation , Polydactyly/metabolism , Polydactyly/pathology , Protein Transport/genetics , Transcription Factors/biosynthesis , Uterine Diseases/metabolism , Uterine Diseases/pathology , Zebrafish/genetics
14.
Biomed Res Int ; 2016: 6437658, 2016.
Article in English | MEDLINE | ID: mdl-27999805

ABSTRACT

The cell-based therapy for cartilage or bone requires a large number of cells; serial passages of chondrocytes are, therefore, needed. However, fates of expanded chondrocytes from extra fingers remain unclarified. The chondrocytes from human epiphyses morphologically changed from small polygonal cells to bipolar elongated spindle cells and to large polygonal cells with degeneration at early passages. Gene of type II collagen was expressed in the cells only at a primary culture (Passage 0) and Passage 1 (P1) cells. The nodules by implantation of P0 to P8 cells were composed of cartilage and perichondrium. The cartilage consisted of chondrocytes with round nuclei and type II collagen-positive matrix, and the perichondrium consisted of spindle cells with type I collage-positive matrix. The cartilage and perichondrium developed to bone with marrow cavity through enchondral ossification. Chondrogenesis and osteogenesis by epiphyseal chondrocytes depended on replication number in culture. It is noteworthy to take population doubling level in correlation with pharmaceutical efficacy into consideration when we use chondrocytes for cell-based therapies.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , Cell- and Tissue-Based Therapy , Chondrocytes/metabolism , Chondrogenesis , Epiphyses/metabolism , Osteogenesis , Cell Nucleus/pathology , Cells, Cultured , Chondrocytes/pathology , Epiphyses/pathology , Female , Humans , Infant , Male , Polydactyly/metabolism , Polydactyly/pathology
15.
Biomed Res Int ; 2015: 861752, 2015.
Article in English | MEDLINE | ID: mdl-26582078

ABSTRACT

Glycosaminoglycans, including chondroitin, dermatan, and heparan sulfate, have various roles in a wide range of biological events such as cell signaling, cell proliferation, tissue morphogenesis, and interactions with various growth factors. Their polysaccharides covalently attach to the serine residues on specific core proteins through the common linker region tetrasaccharide, -xylose-galactose-galactose-glucuronic acid, which is produced through the stepwise addition of respective monosaccharides by four distinct glycosyltransferases. Mutations in the human genes encoding the glycosyltransferases responsible for the biosynthesis of the linker region tetrasaccharide cause a number of genetic disorders, called glycosaminoglycan linkeropathies, including Desbuquois dysplasia type 2, spondyloepimetaphyseal dysplasia, Ehlers-Danlos syndrome, and Larsen syndrome. This review focused on recent studies on genetic diseases caused by defects in the biosynthesis of the common linker region tetrasaccharide.


Subject(s)
Craniofacial Abnormalities/genetics , Dwarfism/genetics , Ehlers-Danlos Syndrome/genetics , Glycosyltransferases/genetics , Joint Instability/genetics , Ossification, Heterotopic/genetics , Osteochondrodysplasias/genetics , Polydactyly/genetics , Cell Proliferation/genetics , Chondroitin/metabolism , Craniofacial Abnormalities/enzymology , Craniofacial Abnormalities/metabolism , Dermatan Sulfate/metabolism , Dwarfism/enzymology , Dwarfism/metabolism , Ehlers-Danlos Syndrome/enzymology , Ehlers-Danlos Syndrome/metabolism , Heparitin Sulfate/metabolism , Humans , Joint Instability/enzymology , Joint Instability/metabolism , Morphogenesis/genetics , Mutation , Ossification, Heterotopic/enzymology , Ossification, Heterotopic/metabolism , Osteochondrodysplasias/enzymology , Osteochondrodysplasias/metabolism , Polydactyly/enzymology , Polydactyly/metabolism
16.
PLoS One ; 10(5): e0128006, 2015.
Article in English | MEDLINE | ID: mdl-26001200

ABSTRACT

The anterior-posterior patterning of the vertebrate limb bud requires closely coordinated signaling interactions, including Sonic Hedgehog (Shh)-mediated counteraction of the Gli3 transcription factor in the distal and posterior mesenchyme of the limb bud. Suppressor of Fused (Sufu), an intracellular negative regulator of Shh signaling via Gli2 and Gli3, is implicated in early development of the mouse limb bud. However, how Sufu is involved in the genetic regulation of limb bud patterning still remains elusive. In this study, we show that the conditional deletion of Sufu in the mesenchyme of the early limb bud results in polydactyly with loss of digit identity and supernumerary bones in the wrist and the ankle. These pattern alterations are associated with anterior expansion of HoxD genes located at the 5' end of the cluster. By focusing on gene expression analysis of Shh/Gremlin1/Fgf signaling critical for the establishment and maintenance of anterior-posterior patterning, we show that early response to loss of Sufu involves anterior prolongation of Fgf4 and Fgf8 expression in the apical ectodermal ridge at E10.5. We also reveal the anterior activation of Shh-dependent posterior markers Ptc1, Gli1 and Gremlin in limb buds lacking Sufu. Furthermore, we find that loss of Sufu leads to attenuated levels of repressor Gli2 and repressor Gli3 in the early limb bud. Moreover, expression of Hand2 is activated in the entire limb bud at the early outgrowth stage in the mutant lacking Sufu. Thus, we provide evidence that Sufu is involved in the genetic network that restricts the posterior expression of Gli2/3/Hand2 and Gremlin/Fgf in limb bud patterning.


Subject(s)
Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction/genetics , Toes/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Body Patterning/genetics , Gene Regulatory Networks , Mice , Mice, Knockout , Polydactyly/genetics , Polydactyly/metabolism , Repressor Proteins/metabolism , Zinc Finger Protein Gli3
17.
Cell Rep ; 9(2): 674-87, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25373905

ABSTRACT

Sorting and degradation of receptors and associated signaling molecules maintain homeostasis of conserved signaling pathways during cell specification and tissue development. Yet, whether machineries that sort signaling proteins act preferentially on different receptors and ligands in different contexts remains mysterious. Here, we show that Vacuolar protein sorting 25, Vps25, a component of ESCRT-II (Endosomal Sorting Complex Required for Transport II), directs preferential endosome-mediated modulation of FGF signaling in limbs. By ENU-induced mutagenesis, we isolated a polydactylous mouse line carrying a hypomorphic mutation of Vps25 (Vps25(ENU)). Unlike Vps25-null embryos we generated, Vps25(ENU/ENU) mutants survive until late gestation. Their limbs display FGF signaling enhancement and consequent hyperactivation of the FGF-SHH feedback loop causing polydactyly, whereas WNT and BMP signaling remain unperturbed. Notably, Vps25(ENU/ENU) Mouse Embryonic Fibroblasts exhibit aberrant FGFR trafficking and degradation; however, SHH signaling is unperturbed. These studies establish that the ESCRT-II machinery selectively limits FGF signaling in vertebrate skeletal patterning.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/metabolism , Polydactyly/genetics , Signal Transduction , Vesicular Transport Proteins/genetics , Animals , Endosomal Sorting Complexes Required for Transport/genetics , Extremities/growth & development , Feedback, Physiological , Fibroblasts/metabolism , Mice , Mice, Inbred C57BL , Mutation , Polydactyly/metabolism , Vesicular Transport Proteins/metabolism
18.
PLoS One ; 8(10): e77311, 2013.
Article in English | MEDLINE | ID: mdl-24204802

ABSTRACT

ETV5 (Ets variant gene 5) is a transcription factor that is required for fertility. In this study, we demonstrate that ETV5 plays additional roles in embryonic and postnatal developmental processes in the mouse. Through a genome-wide mouse mutagenesis approach, we generated a sterile mouse line that carried a nonsense mutation in exon 12 of the Etv5 gene. The mutation led to the conversion of lysine at position 412 into a premature termination codon (PTC) within the ETS DNA binding domain of the protein. We showed that the PTC-containing allele produced a highly unstable mRNA, which in turn resulted in an undetectable level of ETV5 protein. The Etv5 mutation resulted in male and female sterility as determined by breeding experiments. Mutant males were sterile due to a progressive loss of spermatogonia, which ultimately resulted in a Sertoli cell only phenotype by 8 week-of-age. Further, the ETV5 target genes Cxcr4 and Ccl9 were significantly down-regulated in mutant neonate testes. CXCR4 and CCL9 have been implicated in the maintenance and migration of spermatogonia, respectively. Moreover, the Etv5 mutation resulted in several developmental abnormalities including an increased incidence of embryonic and perinatal lethality, postnatal growth restriction, polydactyly and renal asymmetry. Thus, our data define a physiological role for ETV5 in many aspects of development including embryonic and perinatal survival, postnatal growth, limb patterning, kidney development and fertility.


Subject(s)
Body Patterning/genetics , DNA-Binding Proteins/genetics , Infertility/genetics , Mutation, Missense , Polydactyly/genetics , Transcription Factors/genetics , Animals , Chemokines, CC/genetics , Chemokines, CC/metabolism , Codon, Nonsense , DNA-Binding Proteins/metabolism , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Gene Expression Regulation, Developmental , Infertility/metabolism , Infertility/pathology , Kidney/abnormalities , Kidney/metabolism , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , Male , Mice , Mice, Transgenic , Polydactyly/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction , Spermatogenesis/genetics , Spermatogonia/metabolism , Spermatogonia/pathology , Transcription Factors/metabolism
19.
Biochim Biophys Acta ; 1830(3): 2728-38, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23671934

ABSTRACT

BACKGROUND: Upstream open reading frames (uORFs) are commonly found in the 5'-untranslated region (UTR) of many genes and function in translational control. However, little is known about the existence of the proteins encoded by uORFs, and the role of the proteins except translational control. There was no report about uORFs of the McKusick-Kaufman syndrome (MKKS) gene that causes a genetic disorder. METHODS: Northern blotting, 3'-RACE, and bioinformatics were used for determining the length of transcripts and their 3' ends. Luciferase assay and in vitro translation were used for evaluation of translational regulatory activity of uORFs. Immunoblotting and immunocytochemical analyses were used for detection of uORF-derived protein products and their subcellular localization. RESULTS: The MKKS gene generates two types of transcripts: a canonical long transcript that encodes both uORFs and MKKS, and a short transcript that encodes only uORFs by using alternative polyadenylation sites at the 5'-UTR. The simultaneous disruption of the uORF initiation codons increased the translation of the downstream ORF. Furthermore, both protein products from the two longest uORFs were detected in the mitochondrial membrane fraction of HeLa cells. Database searches indicated that such uORFs with active alternative polyadenylation sites at the 5'-UTR are atypical but surely exist in human transcripts. CONCLUSIONS: Multiple uORFs at the 5'-UTR of the MKKS long transcript function as translational repressor for MKKS. Two uORFs are translated in vivo and imported onto the mitochondrial membrane. GENERAL SIGNIFICANCE: Our findings provide unique insights into production of uORF-derived peptides and functions of uORFs.


Subject(s)
5' Untranslated Regions , Abnormalities, Multiple/genetics , Alternative Splicing , Heart Defects, Congenital/genetics , Hydrocolpos/genetics , Mitochondrial Proteins/genetics , Open Reading Frames , Polydactyly/genetics , RNA, Messenger/genetics , Uterine Diseases/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Amino Acid Sequence , Animals , Cell Line, Tumor , Gene Library , Genes, Reporter , Haplorhini , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Humans , Hydrocolpos/metabolism , Hydrocolpos/pathology , Luciferases , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Polyadenylation , Polydactyly/metabolism , Polydactyly/pathology , Protein Biosynthesis , RNA, Messenger/metabolism , Rats , Sequence Alignment , Uterine Diseases/metabolism , Uterine Diseases/pathology
20.
Hum Mutat ; 33(8): 1261-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22539336

ABSTRACT

Desbuquois dysplasia (DD) is characterized by antenatal and postnatal short stature, multiple dislocations, and advanced carpal ossification. Two forms have been distinguished on the basis of the presence (type 1) or the absence (type 2) of characteristic hand anomalies. We have identified mutations in calcium activated nucleotidase 1 gene (CANT1) in DD type 1. Recently, CANT1 mutations have been reported in the Kim variant of DD, characterized by short metacarpals and elongated phalanges. DD has overlapping features with spondyloepiphyseal dysplasia with congenital joint dislocations (SDCD) due to Carbohydrate (chondroitin 6) Sulfotransferase 3 (CHST3) mutations. We screened CANT1 and CHST3 in 38 DD cases (6 type 1 patients, 1 Kim variant, and 31 type 2 patients) and found CANT1 mutations in all DD type 1 cases, the Kim variant and in one atypical DD type 2 expanding the clinical spectrum of hand anomalies observed with CANT1 mutations. We also identified in one DD type 2 case CHST3 mutation supporting the phenotype overlap with SDCD. To further define function of CANT1, we studied proteoglycan synthesis in CANT1 mutated patient fibroblasts, and found significant reduced GAG synthesis in presence of ß-D-xyloside, suggesting that CANT1 plays a role in proteoglycan metabolism.


Subject(s)
Nucleotidases/metabolism , Proteoglycans/metabolism , Cells, Cultured , Chromatography, Gel , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Dwarfism/genetics , Dwarfism/metabolism , Glycosides/metabolism , Humans , Joint Instability/genetics , Joint Instability/metabolism , Mutation , Nucleotidases/genetics , Ossification, Heterotopic/genetics , Ossification, Heterotopic/metabolism , Polydactyly/genetics , Polydactyly/metabolism , Sulfotransferases , Carbohydrate Sulfotransferases
SELECTION OF CITATIONS
SEARCH DETAIL
...