Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Cell ; 179(5): 1207-1221.e22, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31730858

ABSTRACT

Accurate measurement of clonal genotypes, mutational processes, and replication states from individual tumor-cell genomes will facilitate improved understanding of tumor evolution. We have developed DLP+, a scalable single-cell whole-genome sequencing platform implemented using commodity instruments, image-based object recognition, and open source computational methods. Using DLP+, we have generated a resource of 51,926 single-cell genomes and matched cell images from diverse cell types including cell lines, xenografts, and diagnostic samples with limited material. From this resource we have defined variation in mitotic mis-segregation rates across tissue types and genotypes. Analysis of matched genomic and image measurements revealed correlations between cellular morphology and genome ploidy states. Aggregation of cells sharing copy number profiles allowed for calculation of single-nucleotide resolution clonal genotypes and inference of clonal phylogenies and avoided the limitations of bulk deconvolution. Finally, joint analysis over the above features defined clone-specific chromosomal aneuploidy in polyclonal populations.


Subject(s)
DNA Replication/genetics , Genome, Human , High-Throughput Nucleotide Sequencing , Single-Cell Analysis , Aneuploidy , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Shape , Cell Survival , Chromosomes, Human/genetics , Clone Cells , DNA Transposable Elements/genetics , Diploidy , Female , Genotype , Humans , Male , Mice , Mutation/genetics , Phylogeny , Polymorphism, Single Nucleotide/genetics
2.
Development ; 147(13)2020 07 13.
Article in English | MEDLINE | ID: mdl-32554530

ABSTRACT

In mammalian testis, contractile peritubular myoid cells (PMCs) regulate the transport of sperm and luminal fluid, while secreting growth factors and extracellular matrix proteins to support the spermatogonial stem cell niche. However, little is known about the role of testicular smooth muscle cells during postnatal testicular development. Here we report age-dependent expression of hypermethylated in cancer 1 (Hic1; also known as ZBTB29) in testicular smooth muscle cells, including PMCs and vascular smooth muscle cells, in the mouse. Postnatal deletion of Hic1 in smooth muscle cells led to their increased proliferation and resulted in dilatation of seminiferous tubules, with increased numbers of PMCs. These seminiferous tubules contained fewer Sertoli cells and more spermatogonia, and fibronectin was not detected in their basement membrane. The expression levels of genes encoding smooth muscle contractile proteins, Acta2 and Cnn1, were downregulated in the smooth muscle cells lacking Hic1, and the seminiferous tubules appeared to have reduced contractility. These data imply a role for Hic1 in determining the size of seminiferous tubules by regulating postnatal smooth muscle cell proliferation, subsequently affecting spermatogenesis in adulthood.


Subject(s)
Fibronectins/metabolism , Myocytes, Smooth Muscle/metabolism , Testis/metabolism , Actins/genetics , Actins/metabolism , Animals , Fibronectins/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism
3.
Cell Tissue Res ; 389(3): 483-499, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35704103

ABSTRACT

Mesenchymal progenitor cells (MPCs) have been recently identified in human and murine epidural fat and have been hypothesized to contribute to the maintenance/repair/regeneration of the dura mater. MPCs can secrete proteoglycan 4 (PRG4/lubricin), and this protein can regulate tissue homeostasis through bio-lubrication and immunomodulatory functions. MPC lineage tracing reporter mice (Hic1) and human epidural fat MPCs were used to determine if PRG4 is expressed by these cells in vivo. PRG4 expression co-localized with Hic1+ MPCs in the dura throughout skeletal maturity and was localized adjacent to sites of dural injury. When Hic1+ MPCs were ablated, PRG4 expression was retained in the dura, yet when Prx1+ MPCs were ablated, PRG4 expression was completely lost. A number of cellular processes were impacted in human epidural fat MPCs treated with rhPRG4, and human MPCs contributed to the formation of epidural fat, and dura tissues were xenotransplanted into mouse dural injuries. We have shown that human and mouse MPCs in the epidural/dura microenvironment produce PRG4 and can contribute to dura homeostasis/repair/regeneration. Overall, these results suggest that these MPCs have biological significance within the dural microenvironment and that the role of PRG4 needs to be further elucidated.


Subject(s)
Dura Mater/metabolism , Mesenchymal Stem Cells , Proteoglycans/metabolism , Animals , Dura Mater/cytology , Humans , Mesenchymal Stem Cells/metabolism , Mice
4.
Proc Natl Acad Sci U S A ; 116(10): 4518-4527, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30787191

ABSTRACT

Podocalyxin (Podxl) is broadly expressed on the luminal face of most blood vessels in adult vertebrates, yet its function on these cells is poorly defined. In the present study, we identified specific functions for Podxl in maintaining endothelial barrier function. Using electrical cell substrate impedance sensing and live imaging, we found that, in the absence of Podxl, human umbilical vein endothelial cells fail to form an efficient barrier when plated on several extracellular matrix substrates. In addition, these monolayers lack adherens junctions and focal adhesions and display a disorganized cortical actin cytoskeleton. Thus, Podxl has a key role in promoting the appropriate endothelial morphogenesis required to form functional barriers. This conclusion is further supported by analyses of mutant mice in which we conditionally deleted a floxed allele of Podxl in vascular endothelial cells (vECs) using Tie2Cre mice (PodxlΔTie2Cre). Although we did not detect substantially altered permeability in naïve mice, systemic priming with lipopolysaccharide (LPS) selectively disrupted the blood-brain barrier (BBB) in PodxlΔTie2Cre mice. To study the potential consequence of this BBB breach, we used a selective agonist (TFLLR-NH2) of the protease-activated receptor-1 (PAR-1), a thrombin receptor expressed by vECs, neuronal cells, and glial cells. In response to systemic administration of TFLLR-NH2, LPS-primed PodxlΔTie2Cre mice become completely immobilized for a 5-min period, coinciding with severely dampened neuroelectric activity. We conclude that Podxl expression by CNS tissue vECs is essential for BBB maintenance under inflammatory conditions.


Subject(s)
Blood-Brain Barrier , Inflammation/metabolism , Sialoglycoproteins/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Morphogenesis
5.
Genes Dev ; 28(4): 317-27, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24532712

ABSTRACT

Chromatin modulators are emerging as attractive drug targets, given their widespread implication in human cancers and susceptibility to pharmacological inhibition. Here we establish the histone methyltransferase G9a/EHMT2 as a selective regulator of fast proliferating myeloid progenitors with no discernible function in hematopoietic stem cells (HSCs). In mouse models of acute myeloid leukemia (AML), loss of G9a significantly delays disease progression and reduces leukemia stem cell (LSC) frequency. We connect this function of G9a to its methyltransferase activity and its interaction with the leukemogenic transcription factor HoxA9 and provide evidence that primary human AML cells are sensitive to G9A inhibition. Our results highlight a clinical potential of G9A inhibition as a means to counteract the proliferation and self-renewal of AML cells by attenuating HoxA9-dependent transcription.


Subject(s)
Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , HEK293 Cells , Hematopoietic Stem Cells/enzymology , Histone-Lysine N-Methyltransferase/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice, Inbred C57BL , Quinazolines/pharmacology
6.
J Immunol ; 203(12): 3209-3215, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31676672

ABSTRACT

Innate lymphoid cells (ILCs) are critical for host defense and tissue repair but can also contribute to chronic inflammatory diseases. The transcription factor RORα is required for ILC2 development but is also highly expressed by other ILC subsets where its function remains poorly defined. We previously reported that Rorasg/sg bone marrow chimeric mice (C57BL/6J) were protected from Salmonella-induced intestinal fibrosis due to defective ILC3 responses. In this study, single-cell RNA analysis of ILCs isolated from inflamed tissues indicates that RORα perturbation led to a reduction in ILC3 lineages. Furthermore, residual Rorasg/sg ILC3s have decreased expression of key signature genes, including Rorc and activating cytokine receptors. Collectively, our data suggest that RORα plays a key role in preserving functional ILC3s by modulating their ability to integrate environmental cues to efficiently produce cytokines.


Subject(s)
Enteritis/etiology , Enteritis/metabolism , Immunity, Innate , Lymphocytes/immunology , Lymphocytes/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Animals , Biomarkers , Chronic Disease , Disease Models, Animal , Enteritis/pathology , Fibrosis , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice
7.
PLoS Pathog ; 14(2): e1006869, 2018 02.
Article in English | MEDLINE | ID: mdl-29470558

ABSTRACT

The intestinal immune system must be able to respond to a wide variety of infectious organisms while maintaining tolerance to non-pathogenic microbes and food antigens. The Vitamin A metabolite all-trans-retinoic acid (atRA) has been implicated in the regulation of this balance, partially by regulating innate lymphoid cell (ILC) responses in the intestine. However, the molecular mechanisms of atRA-dependent intestinal immunity and homeostasis remain elusive. Here we define a role for the transcriptional repressor Hypermethylated in cancer 1 (HIC1, ZBTB29) in the regulation of ILC responses in the intestine. Intestinal ILCs express HIC1 in a vitamin A-dependent manner. In the absence of HIC1, group 3 ILCs (ILC3s) that produce IL-22 are lost, resulting in increased susceptibility to infection with the bacterial pathogen Citrobacter rodentium. Thus, atRA-dependent expression of HIC1 in ILC3s regulates intestinal homeostasis and protective immunity.


Subject(s)
Immunity, Innate , Intestines/drug effects , Intestines/immunology , Kruppel-Like Transcription Factors/physiology , Lymphocytes/drug effects , Tretinoin/pharmacology , Animals , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/immunology , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Homeostasis/genetics , Homeostasis/immunology , Immunity, Innate/drug effects , Immunity, Innate/genetics , Intestines/microbiology , Kruppel-Like Transcription Factors/genetics , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/drug effects , Signal Transduction/genetics , Tretinoin/metabolism
8.
Development ; 142(24): 4340-50, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26525672

ABSTRACT

Heart valve formation initiates when endothelial cells of the heart transform into mesenchyme and populate the cardiac cushions. The transcription factor SOX9 is highly expressed in the cardiac cushion mesenchyme, and is essential for heart valve development. Loss of Sox9 in mouse cardiac cushion mesenchyme alters cell proliferation, embryonic survival, and valve formation. Despite this important role, little is known about how SOX9 regulates heart valve formation or its transcriptional targets. Therefore, we mapped putative SOX9 binding sites by ChIP-Seq in E12.5 heart valves, a stage at which the valve mesenchyme is actively proliferating and initiating differentiation. Embryonic heart valves have been shown to express a high number of genes that are associated with chondrogenesis, including several extracellular matrix proteins and transcription factors that regulate chondrogenesis. Therefore, we compared regions of putative SOX9 DNA binding between E12.5 heart valves and E12.5 limb buds. We identified context-dependent and context-independent SOX9-interacting regions throughout the genome. Analysis of context-independent SOX9 binding suggests an extensive role for SOX9 across tissues in regulating proliferation-associated genes including key components of the AP-1 complex. Integrative analysis of tissue-specific SOX9-interacting regions and gene expression profiles on Sox9-deficient heart valves demonstrated that SOX9 controls the expression of several transcription factors with previously identified roles in heart valve development, including Twist1, Sox4, Mecom and Pitx2. Together, our data identify SOX9-coordinated transcriptional hierarchies that control cell proliferation and differentiation during valve formation.


Subject(s)
Gene Expression Regulation, Developmental , Heart Valves/embryology , Heart Valves/metabolism , SOX9 Transcription Factor/metabolism , Animals , Cell Proliferation , Chromatin Immunoprecipitation , DNA/metabolism , Extremities/embryology , Gene Regulatory Networks , Mice , Models, Biological , Promoter Regions, Genetic/genetics , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Initiation Site
9.
Hepatology ; 63(6): 1783-95, 2016 06.
Article in English | MEDLINE | ID: mdl-26638120

ABSTRACT

UNLABELLED: Persistent infection of hepatitis C virus (HCV) is one of the leading causes of end-stage liver disease (ESLD), such as decompensated cirrhosis and liver cancer. Of particular note, nearly half of HCV-infected people in the United States are reported to be heavy drinkers. This particular group of patients is known to rapidly progress to the ESLD. Although accelerated disease progression among alcohol abusers infected with HCV is clinically well recognized, the molecular pathophysiology behind this manifestation has not been well elucidated. Hepatocytes metabolize ethanol (EtOH) primarily through two steps of oxidative catabolism in which alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) play central roles. The ADH-ALDH pathway also governs the metabolism of retinol (vitamin A) to its transcriptionally active metabolite, retinoic acid (RA). In this study, we defined that the ADH-ALDH pathway serves as a potent antiviral host factor in hepatocytes, which regulates the expression of interferon (IFN)-stimulated genes (ISGs) by biogenesis of RA. ISGs constitute over 300 antiviral effectors, which cooperatively govern intracellular antiviral innate immunity. Our study revealed that intracellular RA levels greatly influence ISG expression under basal conditions. Moreover, RA augments ISG induction in response to viral infection or exposure to IFN in a gene-specific manner. Lastly, our results demonstrated that EtOH attenuates the antiviral function of the ADH-ALDH pathway, which suggests the possibility that EtOH-retinol metabolic competition is one of the molecular mechanisms for the synergism between HCV and alcohol abuse in liver disease progression. CONCLUSIONS: RA plays a critical role in the regulation of intracellular antiviral innate immunity in hepatocytes. (Hepatology 2016;63:1783-1795).


Subject(s)
Gene Expression Regulation , Hepatocytes/immunology , Immunity, Innate , Liver Failure/etiology , Vitamin A/metabolism , Animals , Cell Line , Ethanol/adverse effects , Ethanol/metabolism , Hepatitis C, Chronic/complications , Hepatocytes/metabolism , Humans , Liver Diseases, Alcoholic/complications , Mice, Inbred C57BL
10.
Fluids Barriers CNS ; 21(1): 31, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38575991

ABSTRACT

BACKGROUND: In the choroid plexus and pituitary gland, vasculature is known to have a permeable, fenestrated phenotype which allows for the free passage of molecules in contrast to the blood brain barrier observed in the rest of the CNS. The endothelium of these compartments, along with secretory, neural-lineage cells (choroid epithelium and pituitary endocrine cells) have been studied in detail, but less attention has been given to the perivascular mesenchymal cells of these compartments. METHODS: The Hic1CreERT2 Rosa26LSL-TdTomato mouse model was used in conjunction with a PdgfraH2B-EGFP mouse model to examine mesenchymal cells, which can be subdivided into Pdgfra+ fibroblasts and Pdgfra- pericytes within the choroid plexus (CP) and pituitary gland (PG), by histological, immunofluorescence staining and single-cell RNA-sequencing analyses. RESULTS: We found that both CP and PG possess substantial populations of distinct Hic1+ mesenchymal cells, including an abundance of Pdgfra+ fibroblasts. Within the pituitary, we identified distinct subpopulations of Hic1+ fibroblasts in the glandular anterior pituitary and the neurosecretory posterior pituitary. We also identified multiple distinct markers of CP, PG, and the meningeal mesenchymal compartment, including alkaline phosphatase, indole-n-methyltransferase and CD34. CONCLUSIONS: Novel, distinct subpopulations of mesenchymal cells can be found in permeable vascular interfaces, including the CP, PG, and meninges, and make distinct contributions to both organs through the production of structural proteins, enzymes, transporters, and trophic molecules.


Subject(s)
Mesenchymal Stem Cells , Red Fluorescent Protein , Mice , Animals , Blood-Brain Barrier/metabolism , Fibroblasts , Single-Cell Analysis , Choroid Plexus/metabolism
11.
bioRxiv ; 2024 May 17.
Article in English | MEDLINE | ID: mdl-38798672

ABSTRACT

Synovial sarcoma (SyS) is an aggressive soft-tissue malignancy characterized by a pathognomonic chromosomal translocation leading to the formation of the SS18::SSX fusion oncoprotein. SS18::SSX associates with mammalian BAF complexes suggesting deregulation of chromatin architecture as the oncogenic driver in this tumour type. To examine the epigenomic state of SyS we performed comprehensive multi-omics analysis on 52 primary pre-treatment human SyS tumours. Our analysis revealed a continuum of epigenomic states across the cohort at fusion target genes independent of rare somatic genetic lesions. We identify cell-of-origin signatures defined by enhancer states and reveal unexpected relationships between H2AK119Ub1 and active marks. The number of bivalent promoters, dually marked by the repressive H3K27me3 and activating H3K4me3 marks, has strong prognostic value and outperforms tumor grade in predicting patient outcome. Finally, we identify SyS defining epigenomic features including H3K4me3 expansion associated with striking promoter DNA hypomethylation in which SyS displays the lowest mean methylation level of any sarcoma subtype. We explore these distinctive features as potential vulnerabilities in SyS and identify H3K4me3 inhibition as a promising therapeutic strategy.

12.
J Cell Sci ; 124(Pt 16): 2723-34, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21807937

ABSTRACT

Cyp26b1, a retinoic acid (RA)-metabolising enzyme, is expressed in the developing limb bud, and Cyp26b1(-/-) mice present with severe limb defects. These malformations might be attributable to an RA-induced patterning defect; however, recent reports suggest that RA is dispensable for limb patterning. In this study, we examined the role of endogenous retinoid signalling in skeletogenesis using Cyp26b1(-/-) mice and transgenic mice in which Cyp26b1 is conditionally deleted under control of the Prrx1 promoter beginning at ~E9.5 (Prrx1Cre(+)/Cyp26b1(fl/fl)). We found that the limb phenotype in Prrx1Cre(+)/Cyp26b1(fl/fl) mice was less severe than that observed in Cyp26b1(-/-) animals and that a change in retinoid signalling contributed to the difference in phenotypes. We systematically examined the role of endogenous RA signalling in chondrogenesis and found that Cyp26b1(-/-) cells and limb mesenchymal cells treated with a CYP inhibitor, are maintained in a pre-chondrogenic state, exhibit reduced chondroblast differentiation and have modestly accelerated chondrocyte hypertrophy. Furthermore, Cyp26b1(-/-) mesenchyme exhibited an increase in expression of genes in a closely related tendogenic lineage, indicating that retinoid signals in the limb interfere with differentiation and maintain progenitor status. Together, these findings support an important function for RA in regulating the behaviour of mesenchymal progenitors, and their subsequent differentiation and maturation.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Extremities/pathology , Mesenchymal Stem Cells/metabolism , Tretinoin/metabolism , Animals , Body Patterning/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cells, Cultured , Chondrogenesis/genetics , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Enzyme Inhibitors/pharmacology , Extremities/embryology , Extremities/growth & development , Homeodomain Proteins/genetics , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mice , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic/genetics , Retinoic Acid 4-Hydroxylase , Signal Transduction/genetics , Transgenes/genetics
13.
Stem Cells ; 30(6): 1152-62, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22415977

ABSTRACT

Pathologies characterized by lipomatous infiltration of craniofacial structures as well as certain forms of lipodystrophies suggest the existence of a distinct adipogenic program in the cephalic region of mammals. Using lineage tracing, we studied the origin of craniofacial adipocytes that accumulate both in cranial fat depots and during ectopic lipomatous infiltration of craniofacial muscles. We found that unlike their counterparts in limb muscle, a significant percentage of cranial adipocytes is derived from the neural crest (NC). In addition, we identified a population of NC-derived Lin(-)/α7(-)/CD34(+)/Sca-1(+) fibro/adipogenic progenitors (NC-FAPs) that resides exclusively in the mesenchyme of cephalic fat and muscle. Comparative analysis of the adipogenic potential, impact on metabolism, and contribution to the regenerative response of NC-FAPs and mesoderm-derived FAPs (M-FAPs) suggests that these cells are functionally indistinguishable. While both NC- and M-FAPs express mesenchymal markers and promyogenic cytokines upon damage-induced activation, NC-FAPs additionally express components of the NC developmental program. Furthermore, we show that craniofacial FAP composition changes with age, with young mice containing FAPs that are almost exclusively of NC origin, while NC-FAPs are progressively replaced by M-FAPs as mice age. Based on these results, we propose that in the adult, ontogenetically distinct FAPs form a diffused system reminiscent of the endothelium, which can originate from multiple developmental intermediates to seed all anatomical locations.


Subject(s)
Adipocytes/physiology , Regeneration/physiology , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Differentiation/physiology , Cell Lineage , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
Cell Rep ; 42(4): 112325, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37002923

ABSTRACT

The musculoskeletal system relies on the integration of multiple components with diverse physical properties, such as striated muscle, tendon, and bone, that enable locomotion and structural stability. This relies on the emergence of specialized, but poorly characterized, interfaces between these various elements during embryonic development. Within the appendicular skeleton, we show that a subset of mesenchymal progenitors (MPs), identified by Hic1, do not contribute to the primary cartilaginous anlagen but represent the MP population, whose progeny directly contribute to the interfaces that connect bone to tendon (entheses), tendon to muscle (myotendinous junctions), and the associated superstructures. Furthermore, deletion of Hic1 leads to skeletal defects reflective of deficient muscle-bone coupling and, consequently, perturbation of ambulation. Collectively, these findings show that Hic1 identifies a unique MP population that contributes to a secondary wave of bone sculpting critical to skeletal morphogenesis.


Subject(s)
Muscle, Skeletal , Tendons , Osteogenesis , Bone and Bones , Cartilage
15.
Sci Rep ; 13(1): 9378, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37296277

ABSTRACT

Promoting bone healing including fracture non-unions are promising targets for bone tissue engineering due to the limited success of current clinical treatment methods. There has been significant research on the use of stem cells with and without biomaterial scaffolds to treat bone fractures due to their promising regenerative capabilities. However, the relative roles of exogenous vs. endogenous stem cells and their overall contribution to in vivo fracture repair is not well understood. The purpose of this study was to determine the interaction between exogenous and endogenous stem cells during bone healing. This study was conducted using a standardized burr-hole bone injury model in a mesenchymal progenitor cell (MPC) lineage-tracing mouse under normal homeostatic and osteoporotic conditions. Burr-hole injuries were treated with a collagen-I biomaterial loaded with and without labelled induced pluripotent stem cells (iPSCs). Using lineage-tracing, the roles of exogenous and endogenous stem cells during bone healing were examined. It was observed that treatment with iPSCs resulted in muted healing compared to untreated controls in intact mice post-injury. When the cell populations were examined histologically, iPSC-treated burr-hole defects presented with a dramatic reduction in endogenous MPCs and cell proliferation throughout the injury site. However, when the ovaries were removed and an osteoporotic-like phenotype induced in the mice, iPSCs treatment resulted in increased bone formation relative to untreated controls. In the absence of iPSCs, endogenous MPCs demonstrated robust proliferative and osteogenic capacity to undertake repair and this behaviour was disrupted in the presence of iPSCs which instead took on an osteoblast fate but with little proliferation. This study clearly demonstrates that exogenously delivered cell populations can impact the normal function of endogenous stem/progenitor populations during the normal healing cascade. These interactions need to be better understood to inform cell and biomaterial therapies to treat fractures.


Subject(s)
Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Mice , Animals , Osteogenesis , Mesenchymal Stem Cells/physiology , Biocompatible Materials , Tissue Engineering/methods , Cell Differentiation
16.
Nat Commun ; 14(1): 5567, 2023 09 09.
Article in English | MEDLINE | ID: mdl-37689753

ABSTRACT

Epithelial-to-mesenchymal transitions (EMTs) of both endocardium and epicardium guide atrioventricular heart valve formation, but the cellular complexity and small scale of this tissue have restricted analyses. To circumvent these issues, we analyzed over 50,000 murine single-cell transcriptomes from embryonic day (E)7.75 hearts to E12.5 atrioventricular canals. We delineate mesenchymal and endocardial bifurcation during endocardial EMT, identify a distinct, transdifferentiating epicardial population during epicardial EMT, and reveal the activation of epithelial-mesenchymal plasticity during both processes. In Sox9-deficient valves, we observe increased epithelial-mesenchymal plasticity, indicating a role for SOX9 in promoting endothelial and mesenchymal cell fate decisions. Lastly, we deconvolve cell interactions guiding the initiation and progression of cardiac valve EMTs. Overall, these data reveal mechanisms of emergence of mesenchyme from endocardium or epicardium at single-cell resolution and will serve as an atlas of EMT initiation and progression with broad implications in regenerative medicine and cancer biology.


Subject(s)
Endocardium , Heart Valves , Animals , Mice , Cell Differentiation , Biology , Cell Communication
17.
Nat Struct Mol Biol ; 30(11): 1640-1652, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37735617

ABSTRACT

The SS18-SSX fusion drives oncogenic transformation in synovial sarcoma by bridging SS18, a member of the mSWI/SNF (BAF) complex, to Polycomb repressive complex 1 (PRC1) target genes. Here we show that the ability of SS18-SSX to occupy H2AK119ub1-rich regions is an intrinsic property of its SSX C terminus, which can be exploited by fusion to transcriptional regulators beyond SS18. Accordingly, SS18-SSX recruitment occurs in a manner that is independent of the core components and catalytic activity of BAF. Alternative SSX fusions are also recruited to H2AK119ub1-rich chromatin and reproduce the expression signatures of SS18-SSX by engaging with transcriptional activators. Variant Polycomb repressive complex 1.1 (PRC1.1) acts as the main depositor of H2AK119ub1 and is therefore required for SS18-SSX occupancy. Importantly, the SSX C terminus not only depends on H2AK119ub1 for localization, but also further increases it by promoting PRC1.1 complex stability. Consequently, high H2AK119ub1 levels are a feature of murine and human synovial sarcomas. These results uncover a critical role for SSX-C in mediating gene deregulation in synovial sarcoma by providing specificity to chromatin and further enabling oncofusion binding by enhancing PRC1.1 stability and H2AK119ub1 deposition.


Subject(s)
Sarcoma, Synovial , Humans , Animals , Mice , Sarcoma, Synovial/genetics , Sarcoma, Synovial/metabolism , Polycomb Repressive Complex 1/genetics , Transcriptional Activation , Cell Nucleus/metabolism , Chromatin/metabolism , Oncogene Proteins, Fusion/metabolism , Cell Cycle Proteins/metabolism
18.
Cancer Res ; 83(21): 3517-3528, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37494476

ABSTRACT

DICER1 is an RNase III enzyme essential for miRNA biogenesis through cleaving precursor-miRNA hairpins. Germline loss-of-function DICER1 mutations underline the development of DICER1 syndrome, a rare genetic disorder that predisposes children to cancer development in organs such as lung, gynecologic tract, kidney, and brain. Unlike classical tumor suppressors, the somatic "second hit" in DICER1 syndrome-associated cancers does not fully inactivate DICER1 but impairs its RNase IIIb activity only, suggesting a noncanonical two-hit hypothesis. Here, we developed a genetically engineered conditional compound heterozygous Dicer1 mutant mouse strain that fully recapitulates the biallelic DICER1 mutations in DICER1 syndrome-associated human cancers. Crossing this tool strain with tissue-specific Cre strains that activate Dicer1 mutations in gynecologic tract cells at two distinct developmental stages revealed that embryonic biallelic Dicer1 mutations caused infertility in females by disrupting oviduct and endometrium development and ultimately drove cancer development. These multicystic tubal and intrauterine tumors histologically resembled a subset of DICER1 syndrome-associated human cancers. Molecular analysis uncovered accumulation of additional oncogenic events (e.g., aberrant p53 expression, Kras mutation, and Myc activation) in murine Dicer1 mutant tumors and validated miRNA biogenesis defects in 5P miRNA strand production, of which, loss of let-7 family miRNAs was identified as a putative key player in transcriptomic rewiring and tumor development. Thus, this DICER1 syndrome-associated cancer model recapitulates the biology of human cancer and provides a unique tool for future investigation and therapeutic development. SIGNIFICANCE: Generation of a Dicer1 mutant mouse model establishes the oncogenicity of missense mutations in the DICER1 RNase IIIb domain and provides a faithful model of DICER1 syndrome-associated cancer for further investigation.


Subject(s)
MicroRNAs , Neoplastic Syndromes, Hereditary , Child , Humans , Female , Animals , Mice , Ribonuclease III/genetics , Ribonuclease III/metabolism , MicroRNAs/genetics , Mutation , Mutation, Missense , DEAD-box RNA Helicases/genetics
19.
Hum Mol Genet ; 19(2): 287-98, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19864492

ABSTRACT

Ocular mal-development results in heterogeneous and frequently visually disabling phenotypes that include coloboma and microphthalmia. Due to the contribution of bone morphogenetic proteins to such processes, the function of the paralogue Growth Differentiation Factor 3 was investigated. Multiple mis-sense variants were identified in patients with ocular and/or skeletal (Klippel-Feil) anomalies including one individual with heterozygous alterations in GDF3 and GDF6. These variants were characterized, individually and in combination, through integrated biochemical and zebrafish model organism analyses, demonstrating appreciable effects with western blot analyses, luciferase based reporter assays and antisense morpholino inhibition. Notably, inhibition of the zebrafish co-orthologue of GDF3 accurately recapitulates patient phenotypes. By demonstrating the pleiotropic effects of GDF3 mutation, these results extend the contribution of perturbed BMP signaling to human disease and potentially implicate multi-allelic inheritance of BMP variants in developmental disorders.


Subject(s)
Eye Abnormalities/genetics , Growth Differentiation Factor 3/genetics , Muscle, Skeletal/abnormalities , Mutation , Amino Acid Sequence , Animals , Cell Line , Eye Abnormalities/metabolism , Female , Growth Differentiation Factor 3/chemistry , Growth Differentiation Factor 3/metabolism , Humans , Male , Molecular Sequence Data , Muscle, Skeletal/metabolism , Pedigree , Sequence Alignment , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
J Cell Sci ; 123(Pt 12): 2068-76, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501701

ABSTRACT

In the developing axial skeleton, sequential sonic hedgehog (SHH) and bone morphogenetic protein (BMP) signals are required for specification of a chondrogenic fate in presomitic tissue. A similar paradigm is thought to operate in the limb, but the signals involved are unclear. To investigate the nature of these signals, we examined BMP action in mesenchymal populations derived from the early murine limb bud (approximately embryonic day 10.5). These populations exhibited a graded response to BMPs, in which early limb mesenchymal cells (from the distal hind limb) displayed an anti-chondrogenic response, whereas BMPs promoted chondrogenesis in more mature cell populations (from the proximal fore limb). Under these conditions, multiple Gata genes were induced by BMPs and the extent of induction correlated with BMP anti-chondrogenic activity. A screen of limb-bud-expressed ligands revealed that prior short-term exposure to transforming growth factor beta1 (TGFbeta1) ameliorated the anti-chondrogenic response to BMP. Furthermore, brief activation of the TGFbeta pathway was found to be necessary for subsequent induction of chondrogenesis by BMPs. Our findings indicate that, similar to axial skeletogenesis, induction of chondrogenesis in the appendicular skeleton is a two-step process. However, the programs differ in the transient signals driving chondrogenic responsiveness to BMPs, with SHH operating in the former and TGFbeta activation in the latter.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Chondrogenesis , Limb Buds/metabolism , Mesoderm/metabolism , Mice/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Cartilage/embryology , Cartilage/metabolism , Cell Differentiation , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Limb Buds/embryology , Male , Mesoderm/embryology , Mice/embryology , Mice/genetics
SELECTION OF CITATIONS
SEARCH DETAIL