Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Sci Transl Med ; 15(724): eabo5217, 2023 11 29.
Article En | MEDLINE | ID: mdl-38019933

Radiotherapy remains a common treatment modality for cancer despite skeletal complications. However, there are currently no effective treatments for radiation-induced bone loss, and the consequences of radiotherapy on skeletal progenitor cell (SPC) survival and function remain unclear. After radiation, leptin receptor-expressing cells, which include a population of SPCs, become localized to hypoxic regions of the bone and stabilize the transcription factor hypoxia-inducible factor-2α (HIF-2α), thus suggesting a role for HIF-2α in the skeletal response to radiation. Here, we conditionally knocked out HIF-2α in leptin receptor-expressing cells and their descendants in mice. Radiation therapy in littermate control mice reduced bone mass; however, HIF-2α conditional knockout mice maintained bone mass comparable to nonirradiated control animals. HIF-2α negatively regulated the number of SPCs, bone formation, and bone mineralization. To test whether blocking HIF-2α pharmacologically could reduce bone loss during radiation, we administered a selective HIF-2α inhibitor called PT2399 (a structural analog of which was recently FDA-approved) to wild-type mice before radiation exposure. Pharmacological inhibition of HIF-2α was sufficient to prevent radiation-induced bone loss in a single-limb irradiation mouse model. Given that ~90% of patients who receive a HIF-2α inhibitor develop anemia because of off-target effects, we developed a bone-targeting nanocarrier formulation to deliver the HIF-2α inhibitor to mouse bone, to increase on-target efficacy and reduce off-target toxicities. Nanocarrier-loaded PT2399 prevented radiation-induced bone loss in mice while reducing drug accumulation in the kidney. Targeted inhibition of HIF-2α may represent a therapeutic approach for protecting bone during radiation therapy.


Basic Helix-Loop-Helix Transcription Factors , Bone Diseases, Metabolic , Humans , Animals , Mice , Basic Helix-Loop-Helix Transcription Factors/physiology , Receptors, Leptin , Mice, Knockout , Stem Cells , Hypoxia-Inducible Factor 1, alpha Subunit
2.
J Bone Miner Res ; 37(5): 983-996, 2022 05.
Article En | MEDLINE | ID: mdl-35220602

Enchondromas and chondrosarcomas are common cartilage neoplasms that are either benign or malignant, respectively. The majority of these tumors harbor mutations in either IDH1 or IDH2. Glutamine metabolism has been implicated as a critical regulator of tumors with IDH mutations. Using genetic and pharmacological approaches, we demonstrated that glutaminase-mediated glutamine metabolism played distinct roles in enchondromas and chondrosarcomas with IDH1 or IDH2 mutations. Glutamine affected cell differentiation and viability in these tumors differently through different downstream metabolites. During murine enchondroma-like lesion development, glutamine-derived α-ketoglutarate promoted hypertrophic chondrocyte differentiation and regulated chondrocyte proliferation. Deletion of glutaminase in chondrocytes with Idh1 mutation increased the number and size of enchondroma-like lesions. In contrast, pharmacological inhibition of glutaminase in chondrosarcoma xenografts reduced overall tumor burden partially because glutamine-derived non-essential amino acids played an important role in preventing cell apoptosis. This study demonstrates that glutamine metabolism plays different roles in tumor initiation and cancer maintenance. Supplementation of α-ketoglutarate and inhibiting GLS may provide a therapeutic approach to suppress enchondroma and chondrosarcoma tumor growth, respectively. © 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).


Bone Neoplasms , Chondroma , Chondrosarcoma , Glutamine , Isocitrate Dehydrogenase , Mutation , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cartilage/metabolism , Chondroma/genetics , Chondroma/metabolism , Chondroma/pathology , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Glutaminase/genetics , Glutaminase/metabolism , Glutamine/genetics , Glutamine/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Ketoglutaric Acids , Mice
3.
JCI Insight ; 6(22)2021 11 22.
Article En | MEDLINE | ID: mdl-34618689

Sarcomas contain a subpopulation of tumor-propagating cells (TPCs) with enhanced tumor-initiating and self-renewal properties. However, it is unclear whether the TPC phenotype in sarcomas is stable or a dynamic cell state that can derive from non-TPCs. In this study, we utilized a mouse model of undifferentiated pleomorphic sarcoma (UPS) to trace the lineage relationship between sarcoma side population (SP) cells that are enriched for TPCs and non-SP cells. By cotransplanting SP and non-SP cells expressing different endogenous fluorescent reporters, we show that non-SP cells can give rise to SP cells with enhanced tumor-propagating potential in vivo. Lineage trajectory analysis using single-cell RNA sequencing from SP and non-SP cells supports the notion that non-SP cells can assume the SP cell phenotype de novo. To test the effect of eradicating SP cells on tumor growth and self-renewal, we generated mouse sarcomas in which the diphtheria toxin receptor is expressed in the SP cells and their progeny. Ablation of the SP population using diphtheria toxin did not impede tumor growth or self-renewal. Altogether, we show that the sarcoma SP represent a dynamic cell state and targeting TPCs alone is insufficient to eliminate tumor progression.


Cell Transformation, Neoplastic/metabolism , Sarcoma/immunology , Side-Population Cells/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Male , Mice , Mice, Inbred NOD , Sarcoma/pathology
4.
Development ; 147(19)2020 10 05.
Article En | MEDLINE | ID: mdl-32907847

Pattern formation is influenced by transcriptional regulation as well as by morphogenetic mechanisms that shape organ primordia, although factors that link these processes remain under-appreciated. Here we show that, apart from their established transcriptional roles in pattern formation, IRX3/5 help to shape the limb bud primordium by promoting the separation and intercalation of dividing mesodermal cells. Surprisingly, IRX3/5 are required for appropriate cell cycle progression and chromatid segregation during mitosis, possibly in a nontranscriptional manner. IRX3/5 associate with, promote the abundance of, and share overlapping functions with co-regulators of cell division such as the cohesin subunits SMC1, SMC3, NIPBL and CUX1. The findings imply that IRX3/5 coordinate early limb bud morphogenesis with skeletal pattern formation.


Chromatids/metabolism , Homeodomain Proteins/metabolism , Limb Buds/embryology , Limb Buds/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Chromosome Segregation/genetics , Chromosome Segregation/physiology , Female , Fluorescent Antibody Technique , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mitosis/genetics , Mitosis/physiology , Pregnancy , RNA-Seq , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics
5.
Nat Cell Biol ; 22(1): 49-59, 2020 01.
Article En | MEDLINE | ID: mdl-31907410

Osteoclasts are multinucleated cells of the monocyte/macrophage lineage that degrade bone. Here, we used lineage tracing studies-labelling cells expressing Cx3cr1, Csf1r or Flt3-to identify the precursors of osteoclasts in mice. We identified an erythromyeloid progenitor (EMP)-derived osteoclast precursor population. Yolk-sac macrophages of EMP origin produced neonatal osteoclasts that can create a space for postnatal bone marrow haematopoiesis. Furthermore, EMPs gave rise to long-lasting osteoclast precursors that contributed to postnatal bone remodelling in both physiological and pathological settings. Our single-cell RNA-sequencing data showed that EMP-derived osteoclast precursors arose independently of the haematopoietic stem cell (HSC) lineage and the data from fate tracking of EMP and HSC lineages indicated the possibility of cell-cell fusion between these two lineages. Cx3cr1+ yolk-sac macrophage descendants resided in the adult spleen, and parabiosis experiments showed that these cells migrated through the bloodstream to the remodelled bone after injury.


Hematopoiesis/physiology , Homeostasis/physiology , Osteoclasts/metabolism , Yolk Sac/metabolism , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Hematopoietic Stem Cells/metabolism , Macrophages/metabolism , Mice
6.
Cell Rep ; 28(11): 2837-2850.e5, 2019 09 10.
Article En | MEDLINE | ID: mdl-31509746

Cellular heterogeneity is frequently observed in cancer, but the biological significance of heterogeneous tumor clones is not well defined. Using multicolor reporters and CRISPR-Cas9 barcoding, we trace clonal dynamics in a mouse model of sarcoma. We show that primary tumor growth is associated with a reduction in clonal heterogeneity. Local recurrence of tumors following surgery or radiation therapy is driven by multiple clones. In contrast, advanced metastasis to the lungs is driven by clonal selection of a single metastatic clone (MC). Using RNA sequencing (RNA-seq) and in vivo assays, we identify candidate suppressors of metastasis, namely, Rasd1, Reck, and Aldh1a2. These genes are downregulated in MCs of the primary tumors prior to the formation of metastases. Overexpression of these suppressors of metastasis impair the ability of sarcoma cells to colonize the lungs. Overall, this study reveals clonal dynamics during each step of tumor progression, from initiation to growth, recurrence, and distant metastasis.


Clonal Evolution/genetics , Clone Cells/metabolism , Neoplasm Recurrence, Local/metabolism , Sarcoma/metabolism , Sarcoma/secondary , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Cell Lineage , Clone Cells/cytology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Luminescent Proteins , Mice , Mice, Nude , Neoplasm Recurrence, Local/genetics , RNA-Seq , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Sarcoma/genetics , Sarcoma/pathology , Transcriptome/genetics , ras Proteins/genetics , ras Proteins/metabolism
7.
Sci Rep ; 9(1): 9005, 2019 06 21.
Article En | MEDLINE | ID: mdl-31227757

ß-catenin protein needs to be precisely regulated for effective fracture repair. The pace of fracture healing slows with age, associated with a transient increase in ß-catenin during the initial phase of the repair process. Here we examined the ability of pharmacologic agents that target ß-catenin to improve the quality of fracture repair in old mice. 20 month old mice were treated with Nefopam or the tankyrase inhibitor XAV939 after a tibia fracture. Fractures were examined 21 days later by micro-CT and histology, and 28 days later using mechanical testing. Daily treatment with Nefopam for three or seven days but not ten days improved the amount of bone present at the fracture site, inhibited ß-catenin protein level, and increased colony forming units osteoblastic from bone marrow cells. At 28 days, treatment increased the work to fracture of the injured tibia. XAV939 had a more modest effect on ß-catenin protein, colony forming units osteoblastic, and the amount of bone at the fracture site. This data supports the notion that high levels of ß-catenin in the early phase of fracture healing in old animals slows osteogenesis, and suggests a pharmacologic approach that targets ß-catenin to improve fracture repair in the elderly.


Fracture Healing/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Nefopam/pharmacology , Tibial Fractures/metabolism , beta Catenin/metabolism , Analgesics, Non-Narcotic/pharmacology , Animals , Male , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Stem Cells/drug effects , Tankyrases/antagonists & inhibitors , Tankyrases/metabolism , Tibia/drug effects , Tibia/injuries , Tibia/metabolism , Tibial Fractures/physiopathology , Time Factors
8.
JCI Insight ; 52019 04 30.
Article En | MEDLINE | ID: mdl-31039139

Enchondroma and chondrosarcoma are the most common benign and malignant cartilaginous neoplasms. Mutations in isocitrate dehydrogenase 1 and 2 (IDH1/2) are present in the majority of these tumors. We performed RNA-seq analysis on chondrocytes from Col2a1Cre;Idh1LSL/+ animals and found that genes implied in cholesterol synthesis pathway were significantly upregulated in the mutant chondrocytes. We examined the phenotypic effect of inhibiting intracellular cholesterol biosynthesis on enchondroma formation by conditionally deleting SCAP (sterol regulatory element-binding protein cleavage-activating protein), a protein activating intracellular cholesterol synthesis, in IDH1 mutant mice. We found fewer enchondromas in animals lacking SCAP. Furthermore, in chondrosarcomas, pharmacological inhibition of intracellular cholesterol synthesis significantly reduced chondrosarcoma cell viability in vitro and suppressed tumor growth in vivo. Taken together, these data suggest that intracellular cholesterol synthesis is a potential therapeutic target for enchondromas and chondrosarcomas.


Cholesterol/biosynthesis , Chondroma/metabolism , Chondrosarcoma/metabolism , Genetic Predisposition to Disease/genetics , Animals , Cell Survival , Chondrocytes/metabolism , Chondroma/drug therapy , Chondroma/genetics , Chondroma/pathology , Chondrosarcoma/drug therapy , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Disease Models, Animal , Isocitrate Dehydrogenase/genetics , Lovastatin/pharmacology , Mice , Mice, Knockout , Xenograft Model Antitumor Assays
9.
Nat Commun ; 9(1): 5191, 2018 12 05.
Article En | MEDLINE | ID: mdl-30518764

The pace of repair declines with age and, while exposure to a young circulation can rejuvenate fracture repair, the cell types and factors responsible for rejuvenation are unknown. Here we report that young macrophage cells produce factors that promote osteoblast differentiation of old bone marrow stromal cells. Heterochronic parabiosis exploiting young mice in which macrophages can be depleted and fractionated bone marrow transplantation experiments show that young macrophages rejuvenate fracture repair, and old macrophage cells slow healing in young mice. Proteomic analysis of the secretomes identify differential proteins secreted between old and young macrophages, such as low-density lipoprotein receptor-related protein 1 (Lrp1). Lrp1 is produced by young cells, and depleting Lrp1 abrogates the ability to rejuvenate fracture repair, while treating old mice with recombinant Lrp1 improves fracture healing. Macrophages and proteins they secrete orchestrate the fracture repair process, and young cells produce proteins that rejuvenate fracture repair in mice.


Fracture Healing , Fractures, Bone/physiopathology , Macrophages/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Female , Fractures, Bone/genetics , Fractures, Bone/metabolism , Fractures, Bone/therapy , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Receptors, LDL/genetics , Rejuvenation , Stromal Cells/cytology , Stromal Cells/metabolism , Stromal Cells/transplantation , Tumor Suppressor Proteins/genetics
10.
Development ; 145(13)2018 07 09.
Article En | MEDLINE | ID: mdl-29899135

During enchondral ossification, mesenchymal cells express genes regulating the intracellular biosynthesis of cholesterol and lipids. Here, we have investigated conditional deletion of Scap or of Insig1 and Insig2 (Scap inhibits intracellular biosynthesis and Insig proteins activate intracellular biosynthesis). Mesenchymal condensation and chondrogenesis was disrupted in mice lacking Scap in mesenchymal progenitors, whereas mice lacking the Insig genes in mesenchymal progenitors had short limbs, but normal chondrogenesis. Mice lacking Scap in chondrocytes showed severe dwarfism, with ectopic hypertrophic cells, whereas deletion of Insig genes in chondrocytes caused a mild dwarfism and shortening of the hypertrophic zone. In vitro studies showed that intracellular cholesterol in chondrocytes can derive from exogenous and endogenous sources, but that exogenous sources cannot completely overcome the phenotypic effect of Scap deficiency. Genes encoding cholesterol biosynthetic proteins are regulated by Hedgehog (Hh) signaling, and Hh signaling is also regulated by intracellular cholesterol in chondrocytes, suggesting a feedback loop in chondrocyte differentiation. Precise regulation of intracellular biosynthesis is required for chondrocyte homeostasis and long bone growth, and these data support pharmacological modulation of cholesterol biosynthesis as a therapy for select cartilage pathologies.


Bone Development/physiology , Cholesterol/biosynthesis , Chondrocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cholesterol/genetics , Chondrocytes/cytology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Signal Transduction/physiology
11.
J Biol Chem ; 293(7): 2466-2475, 2018 02 16.
Article En | MEDLINE | ID: mdl-29196603

Sarcomas, and the mesenchymal precursor cells from which they arise, express chondroitin sulfate proteoglycan 4 (NG2/CSPG4). However, NG2/CSPG4's function and its capacity to serve as a therapeutic target in this tumor type are unknown. Here, we used cells from human tumors and a genetically engineered autochthonous mouse model of soft-tissue sarcomas (STSs) to determine NG2/CSPG4's role in STS initiation and growth. Inhibiting NG2/CSPG4 expression in established murine and human STSs decreased tumor volume by almost two-thirds and cell proliferation rate by 50%. NG2/CSPG4 antibody immunotherapy in human sarcomas established as xenografts in mice similarly decreased tumor volume, and expression of a lentivirus blocking NG2/CSPG4 expression inhibited tumor cell proliferation and increased the latency of engraftment. Gene profiling showed that Ng2/Cspg4 deletion altered the expression of genes regulating cell proliferation and apoptosis. Surprisingly, Ng2/Cspg4 deletion at the time of tumor initiation resulted in larger tumors. Gene expression profiling indicated substantial down-regulation of insulin-like growth factor binding protein (Igfbp) genes when Ng2/Cspg4 is depleted at tumor initiation, but not when Ng2/Cspg4 is depleted after tumor initiation. Such differences may have clinical significance, as therapeutic targeting of a signaling pathway such as NG2/CSPG4 may have different effects on cell behavior with tumor progression. NG2/CSPG4 depletion has divergent effects, depending on the developmental stage of sarcoma. In established tumors, IGF signaling is active, and NG2 inhibition targets cell proliferation and apoptosis.


Antigens/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Membrane Proteins/metabolism , Proteoglycans/metabolism , Sarcoma/metabolism , Sarcoma/physiopathology , Animals , Antigens/genetics , Apoptosis , Cell Line, Tumor , Cell Proliferation , Chondroitin Sulfate Proteoglycans/genetics , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Neoplasm Staging , Proteoglycans/genetics , Sarcoma/genetics , Sarcoma/pathology
12.
J Clin Invest ; 126(5): 1649-63, 2016 05 02.
Article En | MEDLINE | ID: mdl-27018594

Both the WNT/ß-catenin and hedgehog signaling pathways are important in the regulation of limb development, chondrocyte differentiation, and degeneration of articular cartilage in osteoarthritis (OA). It is not clear how these signaling pathways interact in interzone cell differentiation and synovial joint morphogenesis. Here, we determined that constitutive activation of hedgehog signaling specifically within interzone cells induces joint morphological changes by selectively inhibiting ß-catenin-induced Fgf18 expression. Stabilization of ß-catenin or treatment with FGF18 rescued hedgehog-induced phenotypes. Hedgehog signaling induced expression of a dominant negative isoform of TCF7L2 (dnTCF7L2) in interzone progeny, which may account for the selective regulation of ß-catenin target genes observed. Knockdown of TCF7L2 isoforms in mouse chondrocytes rescued hedgehog signaling-induced Fgf18 downregulation, while overexpression of the human dnTCF7L2 orthologue (dnTCF4) in human chondrocytes promoted the expression of catabolic enzymes associated with OA. Similarly, expression of dnTCF4 in human chondrocytes positively correlated with the aggrecanase ADAMTS4. Consistent with our developmental findings, activation of ß-catenin also attenuated hedgehog-induced or surgically induced articular cartilage degeneration in mouse models of OA. Thus, our results demonstrate that hedgehog inhibits selective ß-catenin target gene expression to direct interzone progeny fates and articular cartilage development and disease. Moreover, agents that increase ß-catenin activity have the potential to therapeutically attenuate articular cartilage degeneration as part of OA.


Chondrocytes/immunology , Hedgehog Proteins/immunology , Osteoarthritis/immunology , Synovial Membrane/immunology , Wnt Signaling Pathway/immunology , beta Catenin/immunology , ADAMTS4 Protein/genetics , ADAMTS4 Protein/immunology , Animals , Chondrocytes/pathology , Disease Models, Animal , Down-Regulation/immunology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/immunology , Hedgehog Proteins/genetics , Humans , Mice , Mice, Knockout , Osteoarthritis/genetics , Osteoarthritis/pathology , Synovial Membrane/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factor 7-Like 2 Protein/immunology , Wnt Signaling Pathway/genetics , beta Catenin/genetics
13.
Sci Rep ; 6: 19197, 2016 Jan 20.
Article En | MEDLINE | ID: mdl-26786475

The ventricular conduction system (VCS) orchestrates the harmonious contraction in every heartbeat. Defects in the VCS are often associated with life-threatening arrhythmias and also promote adverse remodeling in heart disease. We have previously established that the Irx3 homeobox gene regulates rapid electrical propagation in the VCS by modulating the transcription of gap junction proteins Cx40 and Cx43. However, it is unknown whether other factors contribute to the conduction defects observed in Irx3 knockout (Irx3(-/-)) mice. In this study, we show that during the early postnatal period, Irx3(-/-) mice develop morphological defects in the VCS which are temporally dissociated from changes in gap junction expression. These morphological defects were accompanied with progressive changes in the cardiac electrocardiogram including right bundle branch block. Hypoplastic VCS was not associated with increased apoptosis of VCS cardiomyocytes but with a lack of recruitment and maturation of ventricular cardiomyocytes into the VCS. Computational analysis followed by functional verification revealed that Irx3 promotes VCS-enriched transcripts targeted by Nkx2.5 and/or Tbx5. Altogether, these results indicate that, in addition to ensuring the appropriate expression of gap junctional channels in the VCS, Irx3 is necessary for the postnatal maturation of the VCS, possibly via its interactions with Tbx5 and Nkx2.5.


Heart Conduction System/growth & development , Heart Conduction System/metabolism , Heart Ventricles/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Cardiac Conduction System Disease , Connexins/genetics , Connexins/metabolism , Electrocardiography , Gene Expression , Gene Expression Regulation , Heart Conduction System/physiopathology , Heart Ventricles/pathology , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Mice , Mice, Knockout , Models, Molecular , Protein Binding , T-Box Domain Proteins/metabolism , Gap Junction alpha-5 Protein
14.
N Engl J Med ; 373(10): 895-907, 2015 Sep 03.
Article En | MEDLINE | ID: mdl-26287746

BACKGROUND: Genomewide association studies can be used to identify disease-relevant genomic regions, but interpretation of the data is challenging. The FTO region harbors the strongest genetic association with obesity, yet the mechanistic basis of this association remains elusive. METHODS: We examined epigenomic data, allelic activity, motif conservation, regulator expression, and gene coexpression patterns, with the aim of dissecting the regulatory circuitry and mechanistic basis of the association between the FTO region and obesity. We validated our predictions with the use of directed perturbations in samples from patients and from mice and with endogenous CRISPR-Cas9 genome editing in samples from patients. RESULTS: Our data indicate that the FTO allele associated with obesity represses mitochondrial thermogenesis in adipocyte precursor cells in a tissue-autonomous manner. The rs1421085 T-to-C single-nucleotide variant disrupts a conserved motif for the ARID5B repressor, which leads to derepression of a potent preadipocyte enhancer and a doubling of IRX3 and IRX5 expression during early adipocyte differentiation. This results in a cell-autonomous developmental shift from energy-dissipating beige (brite) adipocytes to energy-storing white adipocytes, with a reduction in mitochondrial thermogenesis by a factor of 5, as well as an increase in lipid storage. Inhibition of Irx3 in adipose tissue in mice reduced body weight and increased energy dissipation without a change in physical activity or appetite. Knockdown of IRX3 or IRX5 in primary adipocytes from participants with the risk allele restored thermogenesis, increasing it by a factor of 7, and overexpression of these genes had the opposite effect in adipocytes from nonrisk-allele carriers. Repair of the ARID5B motif by CRISPR-Cas9 editing of rs1421085 in primary adipocytes from a patient with the risk allele restored IRX3 and IRX5 repression, activated browning expression programs, and restored thermogenesis, increasing it by a factor of 7. CONCLUSIONS: Our results point to a pathway for adipocyte thermogenesis regulation involving ARID5B, rs1421085, IRX3, and IRX5, which, when manipulated, had pronounced pro-obesity and anti-obesity effects. (Funded by the German Research Center for Environmental Health and others.).


Adipocytes/metabolism , Obesity/genetics , Proteins/genetics , Thermogenesis/genetics , Alleles , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Animals , Base Sequence , Clustered Regularly Interspaced Short Palindromic Repeats , Epigenomics , Gene Expression , Genetic Engineering , Humans , Mice , Mitochondria/metabolism , Molecular Sequence Data , Obesity/metabolism , Phenotype , RNA Editing , Risk , Thermogenesis/physiology
15.
Proc Natl Acad Sci U S A ; 112(9): 2829-34, 2015 Mar 03.
Article En | MEDLINE | ID: mdl-25730874

Enchondromas are benign cartilage tumors and precursors to malignant chondrosarcomas. Somatic mutations in the isocitrate dehydrogenase genes (IDH1 and IDH2) are present in the majority of these tumor types. How these mutations cause enchondromas is unclear. Here, we identified the spectrum of IDH mutations in human enchondromas and chondrosarcomas and studied their effects in mice. A broad range of mutations was identified, including the previously unreported IDH1-R132Q mutation. These mutations harbored enzymatic activity to catalyze α-ketoglutarate to d-2-hydroxyglutarate (d-2HG). Mice expressing Idh1-R132Q in one allele in cells expressing type 2 collagen showed a disordered growth plate, with persistence of type X-expressing chondrocytes. Chondrocyte cell cultures from these animals or controls showed that there was an increase in proliferation and expression of genes characteristic of hypertrophic chondrocytes with expression of Idh1-R132Q or 2HG treatment. Col2a1-Cre;Idh1-R132Q mutant knock-in mice (mutant allele expressed in chondrocytes) did not survive after the neonatal stage. Col2a1-Cre/ERT2;Idh1-R132 mutant conditional knock-in mice, in which Cre was induced by tamoxifen after weaning, developed multiple enchondroma-like lesions. Taken together, these data show that mutant IDH or d-2HG causes persistence of chondrocytes, giving rise to rests of growth-plate cells that persist in the bone as enchondromas.


Chondrocytes , Enchondromatosis , Gene Expression Regulation, Enzymologic , Isocitrate Dehydrogenase , Mutation, Missense , Amino Acid Substitution , Animals , Chondrocytes/enzymology , Chondrocytes/pathology , Collagen Type II/biosynthesis , Collagen Type II/genetics , Enchondromatosis/enzymology , Enchondromatosis/genetics , Enchondromatosis/pathology , Glutarates/adverse effects , Glutarates/pharmacology , Humans , Isocitrate Dehydrogenase/biosynthesis , Isocitrate Dehydrogenase/genetics , Mice , Mice, Mutant Strains
16.
PLoS One ; 10(3): e0119455, 2015.
Article En | MEDLINE | ID: mdl-25760946

Hedgehog signaling is primarily transduced by two transcription factors: Gli2, which mainly acts as a full-length activator, and Gli3, which tends to be proteolytically processed from a full-length form (Gli3FL) to an N-terminal repressor (Gli3REP). Recent studies using a Sufu knockout mouse have indicated that Sufu is involved in regulating Gli2 and Gli3 activator and repressor activity at multiple steps of the signaling cascade; however, the mechanism of specific Gli2 and Gli3 regulation remains to be elucidated. In this study, we established an allelic series of ENU-induced mouse strains. Analysis of one of the missense alleles, SufuT396I, showed that Thr396 residue of Sufu played a key role in regulation of Gli3 activity. SufuT396I/T396I embryos exhibited severe polydactyly, which is indicative of compromised Gli3 activity. Concomitantly, significant quantitative reductions of unprocessed Gli3 (Gli3FL) and processed Gli3 (Gli3REP) were observed in vivo as well as in vitro. Genetic experiments showed that patterning defects in the limb buds of SufuT396I/T396I were rescued by a constitutive Gli3REP allele (Gli3∆699), strongly suggesting that SufuT396I reduced the truncated Gli3 repressor. In contrast, SufuT396I qualitatively exhibited no mutational effects on Gli2 regulation. Taken together, the results of this study show that the Thr396 residue of Sufu is specifically required for regulation of Gli3 but not Gli2. This implies a novel Sufu-mediated mechanism in which Gli2 activator and Gli3 repressor are differentially regulated.


Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/metabolism , Mutation, Missense , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Body Patterning , Extremities/growth & development , Isoleucine/metabolism , Mice , Polydactyly/embryology , Polydactyly/genetics , Protein Stability , Threonine/metabolism , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
17.
Sci Signal ; 7(355): ra117, 2014 Dec 09.
Article En | MEDLINE | ID: mdl-25492966

The primary cilium is required for Hedgehog (Hh) signaling in vertebrates. Hh leads to ciliary accumulation and activation of the transmembrane protein Smoothened (Smo) and affects the localization of several pathway components, including the Gli family of transcriptional regulators, within different regions of primary cilia. Genetic analysis indicates that the kinesin protein Kif7 both promotes and inhibits mouse Hh signaling. Using mass spectrometry, we identified liprin-α1 (PPFIA1) and the protein phosphatase PP2A as Kif7-interacting proteins, and we showed that they were important for the trafficking of Kif7 and Gli proteins to the tips of cilia and for the transcriptional output of Hh signaling. Our results suggested that PPFIA1 functioned with PP2A to promote the dephosphorylation of Kif7, triggering Kif7 localization to the tips of primary cilia and promoting Gli transcriptional activity.


Adaptor Proteins, Signal Transducing/metabolism , Hedgehog Proteins/metabolism , Kinesins/metabolism , Protein Phosphatase 2/metabolism , Proteins/metabolism , Signal Transduction/physiology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cilia/genetics , Cilia/metabolism , HEK293 Cells , Hedgehog Proteins/genetics , Humans , Kinesins/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Protein Phosphatase 2/genetics , Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/physiology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zinc Finger Protein GLI1
18.
Dev Cell ; 29(2): 233-40, 2014 Apr 28.
Article En | MEDLINE | ID: mdl-24726282

Limb skeletal pattern relies heavily on graded Sonic hedgehog (Shh) signaling. As a morphogen and growth cue, Shh regulates identities of posterior limb elements, including the ulna/fibula and digits 2 through 5. In contrast, proximal and anterior structures, including the humerus/femur, radius/tibia, and digit 1, are regarded as Shh independent, and mechanisms governing their specification are unclear. Here, we show that patterning of the proximal and anterior limb skeleton involves two phases. Irx3 and Irx5 (Irx3/5) are essential in the initiating limb bud to specify progenitors of the femur, tibia, and digit 1. However, these skeletal elements can be restored in Irx3/5 null mice when Shh signaling is diminished, indicating that Shh negatively regulates their formation after initiation. Our data provide genetic evidence supporting the concept of early specification and progressive determination of anterior limb pattern.


Bone Development/physiology , Hedgehog Proteins/metabolism , Homeodomain Proteins/metabolism , Limb Buds/embryology , Limb Buds/metabolism , Transcription Factors/metabolism , Animals , Body Patterning/physiology , Femur/embryology , Femur/physiology , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hindlimb/embryology , Hindlimb/physiology , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mutation , Signal Transduction/physiology , Tibia/embryology , Tibia/physiology , Toes/embryology , Toes/physiology , Transcription Factors/genetics
19.
Dev Cell ; 29(2): 241-9, 2014 Apr 28.
Article En | MEDLINE | ID: mdl-24726283

The patterning and growth of the embryonic vertebrate limb is dependent on Sonic hedgehog (Shh), a morphogen that regulates the activity of Gli transcription factors. However, Shh expression is not observed during the first 12 hr of limb development. During this phase, the limb bud is prepatterned into anterior and posterior regions through the antagonistic actions of transcription factors Gli3 and Hand2. We demonstrate that precocious activation of Shh signaling during this early phase interferes with the Gli3-dependent specification of anterior progenitors, disturbing establishment of signaling centers and normal outgrowth of the limb. Our findings illustrate that limb development requires a sweet spot in the level and timing of pathway activation that allows for the Shh-dependent expansion of posterior progenitors without interfering with early prepatterning functions of Gli3/Gli3R or specification of anterior progenitors.


Hedgehog Proteins/metabolism , Limb Buds/embryology , Limb Buds/metabolism , Signal Transduction/physiology , Animals , Animals, Outbred Strains , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/physiology , Cattle , Chickens , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Kinesins/genetics , Kinesins/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Zinc Finger Protein Gli3
20.
Nature ; 507(7492): 371-5, 2014 Mar 20.
Article En | MEDLINE | ID: mdl-24646999

Genome-wide association studies (GWAS) have reproducibly associated variants within introns of FTO with increased risk for obesity and type 2 diabetes (T2D). Although the molecular mechanisms linking these noncoding variants with obesity are not immediately obvious, subsequent studies in mice demonstrated that FTO expression levels influence body mass and composition phenotypes. However, no direct connection between the obesity-associated variants and FTO expression or function has been made. Here we show that the obesity-associated noncoding sequences within FTO are functionally connected, at megabase distances, with the homeobox gene IRX3. The obesity-associated FTO region directly interacts with the promoters of IRX3 as well as FTO in the human, mouse and zebrafish genomes. Furthermore, long-range enhancers within this region recapitulate aspects of IRX3 expression, suggesting that the obesity-associated interval belongs to the regulatory landscape of IRX3. Consistent with this, obesity-associated single nucleotide polymorphisms are associated with expression of IRX3, but not FTO, in human brains. A direct link between IRX3 expression and regulation of body mass and composition is demonstrated by a reduction in body weight of 25 to 30% in Irx3-deficient mice, primarily through the loss of fat mass and increase in basal metabolic rate with browning of white adipose tissue. Finally, hypothalamic expression of a dominant-negative form of Irx3 reproduces the metabolic phenotypes of Irx3-deficient mice. Our data suggest that IRX3 is a functional long-range target of obesity-associated variants within FTO and represents a novel determinant of body mass and composition.


Homeodomain Proteins/genetics , Introns/genetics , Mixed Function Oxygenases/genetics , Obesity/genetics , Oxo-Acid-Lyases/genetics , Proteins/genetics , Transcription Factors/genetics , Adipose Tissue/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Animals , Basal Metabolism/genetics , Body Mass Index , Body Weight/genetics , Brain/metabolism , Diabetes Mellitus, Type 2/genetics , Diet , Genes, Dominant/genetics , Homeodomain Proteins/metabolism , Humans , Hypothalamus/metabolism , Male , Mice , Phenotype , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Thinness/genetics , Transcription Factors/deficiency , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/genetics
...