Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters











Publication year range
1.
Hepatol Commun ; 5(12): 2019-2034, 2021 12.
Article in English | MEDLINE | ID: mdl-34558852

ABSTRACT

We previously identified an up-regulation of specific Wnt proteins in the cholangiocyte compartment during cholestatic liver injury and found that mice lacking Wnt secretion from hepatocytes and cholangiocytes showed fewer proliferating cholangiocytes and high mortality in response to a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet, a murine model of primary sclerosing cholangitis. In vitro studies demonstrated that Wnt7b, one of the Wnts up-regulated during cholestasis, induces proliferation of cholangiocytes in an autocrine manner and increases secretion of proinflammatory cytokines. We hypothesized that loss of Wnt7b may exacerbate some of the complications of cholangiopathies by decreasing the ability of bile ducts to induce repair. Wnt7b-flox mice were bred with Krt19-cre mice to deplete Wnt7b expression in only cholangiocytes (CC) or with albumin-Cre mice to delete Wnt7b expression in both hepatocytes and cholangiocytes (HC + CC). These mice were placed on a DDC diet for 1 month then killed for evaluation. Contrary to our expectations, we found that mice lacking Wnt7b from CC and HC + CC compartments had improved biliary injury, decreased cellular senescence, and lesser bile acid accumulation after DDC exposure compared to controls, along with decreased expression of inflammatory cytokines. Although Wnt7b knockout (KO) resulted in fewer proliferating cholangiocytes, CC and HC + CC KO mice on a DDC diet also had more hepatocytes expressing cholangiocyte markers compared to wild-type mice on a DDC diet, indicating that Wnt7b suppression promotes hepatocyte reprogramming. Conclusion: Wnt7b induces a proproliferative proinflammatory program in cholangiocytes, and its loss is compensated for by conversion of hepatocytes to a biliary phenotype during cholestatic injury.


Subject(s)
Bile Ducts/cytology , Cell Proliferation/genetics , Cholestasis/genetics , Proto-Oncogene Proteins/deficiency , Wnt Proteins/deficiency , Animals , Bile Acids and Salts/metabolism , Cellular Senescence/genetics , Disease Models, Animal , Hepatocytes/metabolism , Mice , Mice, Knockout
2.
J Clin Invest ; 131(16)2021 08 16.
Article in English | MEDLINE | ID: mdl-34255743

ABSTRACT

In view of emerging drug-resistant tuberculosis (TB), host-directed adjunct therapies are urgently needed to improve treatment outcomes with currently available anti-TB therapies. One approach is to interfere with the formation of lipid-laden "foamy" macrophages in the host, as they provide a nutrient-rich host cell environment for Mycobacterium tuberculosis (Mtb). Here, we provide evidence that Wnt family member 6 (WNT6), a ligand of the evolutionarily conserved Wingless/Integrase 1 (WNT) signaling pathway, promotes foam cell formation by regulating key lipid metabolic genes including acetyl-CoA carboxylase 2 (ACC2) during pulmonary TB. Using genetic and pharmacological approaches, we demonstrated that lack of functional WNT6 or ACC2 significantly reduced intracellular triacylglycerol (TAG) levels and Mtb survival in macrophages. Moreover, treatment of Mtb-infected mice with a combination of a pharmacological ACC2 inhibitor and the anti-TB drug isoniazid (INH) reduced lung TAG and cytokine levels, as well as lung weights, compared with treatment with INH alone. This combination also reduced Mtb bacterial numbers and the size of mononuclear cell infiltrates in livers of infected mice. In summary, our findings demonstrate that Mtb exploits WNT6/ACC2-induced storage of TAGs in macrophages to facilitate its intracellular survival, a finding that opens new perspectives for host-directed adjunctive treatment of pulmonary TB.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Proto-Oncogene Proteins/metabolism , Triglycerides/metabolism , Wnt Proteins/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Animals , Antitubercular Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Foam Cells/metabolism , Host Microbial Interactions/drug effects , Host Microbial Interactions/physiology , Humans , Isoniazid/administration & dosage , Lung/drug effects , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/drug effects , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/microbiology , Wnt Proteins/deficiency , Wnt Proteins/genetics
3.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34206401

ABSTRACT

Wingless-type MMTV integration site family, member 16 (wnt16), is a wnt ligand that participates in the regulation of vertebrate skeletal development. Studies have shown that wnt16 can regulate bone metabolism, but its molecular mechanism remains largely undefined. We obtained the wnt16-/- zebrafish model using the CRISPR-Cas9-mediated gene knockout screen with 11 bp deletion in wnt16, which led to the premature termination of amino acid translation and significantly reduced wnt16 expression, thus obtaining the wnt16-/- zebrafish model. The expression of wnt16 in bone-related parts was detected via in situ hybridization. The head, spine, and tail exhibited significant deformities, and the bone mineral density and trabecular bone decreased in wnt16-/- using light microscopy and micro-CT analysis. RNA sequencing was performed to explore the differentially expressed genes (DEGs). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis found that the down-regulated DEGs are mainly concentrated in mTOR, FoxO, and VEGF pathways. Protein-protein interaction (PPI) network analysis was performed with the detected DEGs. Eight down-regulated DEGs including akt1, bnip4, ptena, vegfaa, twsg1b, prkab1a, prkab1b, and pla2g4f.2 were validated by qRT-PCR and the results were consistent with the RNA-seq data. Overall, our work provides key insights into the influence of wnt16 gene on skeletal development.


Subject(s)
Bone and Bones/abnormalities , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/metabolism , Osteogenesis/genetics , Wnt Proteins/deficiency , Zebrafish Proteins/deficiency , Zebrafish/genetics , Animals , Animals, Genetically Modified , Computational Biology/methods , Disease Models, Animal , Gene Expression Profiling , Gene Knockout Techniques , Gene Ontology , Molecular Sequence Annotation , Musculoskeletal Abnormalities/diagnosis , Phenotype , Transcriptome , Wnt Proteins/chemistry , Wnt Proteins/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
4.
J Cereb Blood Flow Metab ; 41(8): 2132-2133, 2021 08.
Article in English | MEDLINE | ID: mdl-33969732

ABSTRACT

For efficient stroke recovery, the entire neurovascular unit must be repaired. A recent study underscores this concept by highlighting the importance of cellular crosstalk for white mater remodeling. In developing brains and in brains injured by hypoxia, interactions between oligodendrocyte precursors and endothelium play an essential role for physiological and compensatory angiogenesis. Further studies are warranted to build on these emerging findings in the oligovascular niche in order to identify novel therapeutic targets for stroke and other CNS diseases.


Subject(s)
Neovascularization, Physiologic , Oligodendrocyte Precursor Cells/metabolism , Animals , Cell Communication , Mice , Mice, Knockout , Oligodendrocyte Precursor Cells/cytology , White Matter/blood supply , White Matter/growth & development , Wnt Proteins/deficiency , Wnt Proteins/genetics
5.
Ann Rheum Dis ; 78(4): 551-561, 2019 04.
Article in English | MEDLINE | ID: mdl-30745310

ABSTRACT

OBJECTIVES: Wnt16 is implicated in bone fracture and bone mass accrual both in animals and humans. However, its functional roles and molecular mechanism in chondrocyte differentiation and osteoarthritis (OA) pathophysiology remain largely undefined. In this study, we analysed its mechanistic association and functional relationship in OA progression in chondrocyte lineage. METHODS: The role of Wnt16 during skeletal development was examined by Col2a1-Wnt16 transgenic mice and Wnt16fl/fl;Col2a1-Cre (Wnt16-cKO) mice. OA progression was assessed by micro-CT analysis and Osteoarthritis Research Society International score after anterior cruciate ligament transection (ACLT) surgery with Wnt16 manipulation by adenovirus intra-articular injection. The molecular mechanism was investigated in vitro using 3D chondrocyte pellet culture and biochemical analyses. Histological analysis was performed in mouse joints and human cartilage specimens. RESULTS: Wnt16 overexpression in chondrocytes in mice significantly inhibited chondrocyte hypertrophy during skeletal development. Wnt16 deficiency exaggerated OA progression, whereas intra-articular injection of Ad-Wnt16 markedly attenuated ACLT-induced OA. Cellular and molecular analyses showed that, instead of ß-catenin and calcium pathways, Wnt16 activated the planar cell polarity (PCP) and JNK pathway by interacting mainly with AP2b1, and to a lesser extend Ror2 and CD146, and subsequently induced PTHrP expression through phosphor-Raptor mTORC1 pathway. CONCLUSIONS: Our findings indicate that Wnt16 activates PCP/JNK and crosstalks with mTORC1-PTHrP pathway to inhibit chondrocyte hypertrophy. Our preclinical study suggests that Wnt16 may be a potential therapeutic target for OA treatment.


Subject(s)
Arthritis, Experimental/pathology , Osteoarthritis/pathology , Wnt Proteins/physiology , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/physiopathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Differentiation/physiology , Cell Polarity/physiology , Cell Proliferation/physiology , Cells, Cultured , Chondrocytes/pathology , Chondrocytes/physiology , Disease Progression , Humans , Hypertrophy/prevention & control , MAP Kinase Signaling System/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Mice, Transgenic , Osteoarthritis/metabolism , Osteoarthritis/physiopathology , Parathyroid Hormone-Related Protein/physiology , Wnt Proteins/deficiency , Wnt Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 115(50): E11827-E11836, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30478038

ABSTRACT

ß-Catenin signaling controls the development and maintenance of the blood-brain barrier (BBB) and the blood-retina barrier (BRB), but the division of labor and degree of redundancy between the two principal ligand-receptor systems-the Norrin and Wnt7a/Wnt7b systems-are incompletely defined. Here, we present a loss-of-function genetic analysis of postnatal BBB and BRB maintenance in mice that shows striking threshold and partial redundancy effects. In particular, the combined loss of Wnt7a and Norrin or Wnt7a and Frizzled4 (Fz4) leads to anatomically localized BBB defects that are far more severe than observed with loss of Wnt7a, Norrin, or Fz4 alone. In the cerebellum, selective loss of Wnt7a in glia combined with ubiquitous loss of Norrin recapitulates the phenotype observed with ubiquitous loss of both Wnt7a and Norrin, implying that glia are the source of Wnt7a in the cerebellum. Tspan12, a coactivator of Norrin signaling in the retina, is also active in BBB maintenance but is less potent than Norrin, consistent with a model in which Tspan12 enhances the amplitude of the Norrin signal in vascular endothelial cells. Finally, in the context of a partially impaired Norrin system, the retina reveals a small contribution to BRB development from the Wnt7a/Wnt7b system. Taken together, these experiments define the extent of CNS region-specific cooperation for several components of the Norrin and Wnt7a/Wnt7b systems, and they reveal substantial regional heterogeneity in the extent to which partially redundant ligands, receptors, and coactivators maintain the BBB and BRB.


Subject(s)
Blood-Brain Barrier/growth & development , Blood-Brain Barrier/physiology , Blood-Retinal Barrier/growth & development , Blood-Retinal Barrier/physiology , Eye Proteins/physiology , Nerve Tissue Proteins/physiology , Proto-Oncogene Proteins/physiology , Wnt Proteins/physiology , Animals , Blood-Brain Barrier/cytology , Blood-Retinal Barrier/cytology , Cell Culture Techniques , Eye Proteins/genetics , Frizzled Receptors/deficiency , Frizzled Receptors/genetics , Frizzled Receptors/physiology , Mice , Mice, Knockout , Models, Biological , Models, Neurological , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Signal Transduction , Tetraspanins/deficiency , Tetraspanins/genetics , Tetraspanins/physiology , Wnt Proteins/deficiency , Wnt Proteins/genetics , beta Catenin/physiology
7.
Ann N Y Acad Sci ; 1415(1): 21-33, 2018 03.
Article in English | MEDLINE | ID: mdl-29500936

ABSTRACT

Activated lymphocytes promote inflammation and bone destruction in rheumatoid arthritis (RA), making T cells and B cells therapeutic targets. Indeed, pharmacological blockade of CD28 costimulation using CTLA-4Ig (abatacept), approved for amelioration of RA, renders T cells dormant (anergic). CTLA-4Ig also promotes bone accretion in healthy mice; surprisingly, however, this effect is driven exclusively through upregulation of bone formation, rather than anti-inflammatory effects on resorption. In the study presented here, we utilized T cell receptor ß gene and Wnt-10b gene knockout mice to investigate the roles of T cells and Wnt-10b in CTLA-4Ig-induced bone anabolism. Ablation of either T cells or Wnt-10b not only abolished CTLA-4Ig-induced bone anabolism but also, paradoxically, suppressed bone formation leading to bone loss. Stalled bone formation was accompanied by bone marrow stromal cell expression of the Wnt pathway inhibitor sclerostin. Our data suggest that an immunoskeletal pivot may promote or suppress bone formation, depending on the net outcome of CTLA-4Ig action directed independently on T cells and osteoblast-linage cells that counter Wnt-10b-induced bone anabolism, by secretion of sclerostin. While CTLA-4Ig action is tipped in favor of bone formation under physiological conditions, pathological immunodeficiency may lead to suppressed bone formation and skeletal damage.


Subject(s)
Abatacept/pharmacology , Anabolic Agents/pharmacology , Bone and Bones/drug effects , Glycoproteins/metabolism , T-Lymphocytes/drug effects , Wnt Proteins/metabolism , 3T3 Cells , Adaptor Proteins, Signal Transducing , Animals , Antirheumatic Agents/pharmacology , Bone Density/drug effects , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , CD28 Antigens/metabolism , Female , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Wnt Proteins/deficiency , Wnt Proteins/genetics , X-Ray Microtomography
8.
Ann Rheum Dis ; 76(1): 218-226, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27147711

ABSTRACT

OBJECTIVE: Both excessive and insufficient activation of WNT signalling results in cartilage breakdown and osteoarthritis. WNT16 is upregulated in the articular cartilage following injury and in osteoarthritis. Here, we investigate the function of WNT16 in osteoarthritis and the downstream molecular mechanisms. METHODS: Osteoarthritis was induced by destabilisation of the medial meniscus in wild-type and WNT16-deficient mice. Molecular mechanisms and downstream effects were studied in vitro and in vivo in primary cartilage progenitor cells and primary chondrocytes. The pathway downstream of WNT16 was studied in primary chondrocytes and using the axis duplication assay in Xenopus. RESULTS: WNT16-deficient mice developed more severe osteoarthritis with reduced expression of lubricin and increased chondrocyte apoptosis. WNT16 supported the phenotype of cartilage superficial-zone progenitor cells and lubricin expression. Increased osteoarthritis in WNT16-deficient mice was associated with excessive activation of canonical WNT signalling. In vitro, high doses of WNT16 weakly activated canonical WNT signalling, but, in co-stimulation experiments, WNT16 reduced the capacity of WNT3a to activate the canonical WNT pathway. In vivo, WNT16 rescued the WNT8-induced primary axis duplication in Xenopus embryos. CONCLUSIONS: In osteoarthritis, WNT16 maintains a balanced canonical WNT signalling and prevents detrimental excessive activation, thereby supporting the homeostasis of progenitor cells.


Subject(s)
Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Wnt Proteins/physiology , Wnt Signaling Pathway/physiology , Animals , Apoptosis/physiology , Arthritis, Experimental/etiology , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Male , Mice, Knockout , Osteoarthritis/etiology , Osteoarthritis/pathology , Proteoglycans/biosynthesis , Proteoglycans/genetics , RNA, Messenger/genetics , Up-Regulation/physiology , Wnt Proteins/biosynthesis , Wnt Proteins/deficiency , Wnt Proteins/genetics
9.
BMC Med Genet ; 17(1): 88, 2016 Nov 24.
Article in English | MEDLINE | ID: mdl-27881089

ABSTRACT

BACKGROUND: The WNT10A protein is critical for the development of ectodermal appendages. Variants in the WNT10A gene may be associated with a spectrum of ectodermal abnormalities including extensive tooth agenesis. METHODS: In seven patients with severe tooth agenesis we identified anomalies in primary dentition and additional ectodermal symptoms, and assessed WNT10A mutations by genetic analysis. RESULTS: Investigation of primary dentition revealed peg-shaped crowns of primary mandibular incisors and three individuals had agenesis of at least two primary teeth. The permanent dentition was severely affected in all individuals with a mean of 21 missing teeth. Primary teeth were most often present in positions were succedaneous teeth were missing. Furthermore, most existing molars had taurodontism. Light, brittle or coarse hair was reported in all seven individuals, hyperhidrosis of palms and soles in six individuals and nail anomalies in two individuals. The anomalies in primary dentition preceded most of the additional ectodermal symptoms. Genetic analysis revealed that all seven individuals were homozygous or compound heterozygous for WNT10A mutations resulting in C107X, E222X and F228I. CONCLUSIONS: We conclude that tooth agenesis and/or peg-shaped crowns of primary mandibular incisors, severe oligodontia of permanent dentition as well as ectodermal symptoms of varying severity may be predictors of bi-allelic WNT10A mutations of importance for diagnosis, counselling and follow-up.


Subject(s)
Ectodermal Dysplasia/genetics , Mutation , Tooth Abnormalities/genetics , Wnt Proteins/genetics , Adolescent , Anodontia/genetics , Child , Dental Enamel Hypoplasia/genetics , Dentition, Permanent , Female , Homozygote , Humans , Male , Tooth, Deciduous/abnormalities , Wnt Proteins/deficiency
10.
Sci Rep ; 6: 19223, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26771085

ABSTRACT

Transcriptional and signaling networks establish complex cross-regulatory interactions that drive cellular differentiation during development. Using microarrays we identified the gene encoding the ligand Wnt9a as a candidate target of Neurogenin3, a basic helix-loop-helix transcription factor that functions as a master regulator of pancreatic endocrine differentiation. Here we show that Wnt9a is expressed in the embryonic pancreas and that its deficiency enhances activation of the endocrine transcriptional program and increases the number of endocrine cells at birth. We identify the gene encoding the endocrine transcription factor Nkx2-2 as one of the most upregulated genes in Wnt9a-ablated pancreases and associate its activation to reduced expression of the Wnt effector Tcf7l2. Accordingly, in vitro studies confirm that Tcf7l2 represses activation of Nkx2-2 by Neurogenin3 and inhibits Nkx2-2 expression in differentiated ß-cells. Further, we report that Tcf7l2 protein levels decline upon initiation of endocrine differentiation in vivo, disclosing the downregulation of this factor in the developing endocrine compartment. These findings highlight the notion that modulation of signalling cues by lineage-promoting factors is pivotal for controlling differentiation programs.


Subject(s)
Organogenesis , Pancreas/embryology , Pancreas/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Wnt Proteins/deficiency , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Endocrine Cells/metabolism , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Mice , Models, Biological , Nerve Tissue Proteins/metabolism , Organogenesis/genetics , Pancreas/anatomy & histology , Pancreas/cytology , Phenotype , Signal Transduction , Transcription Factor 7-Like 2 Protein/genetics , Transcription Factors/genetics , Zebrafish Proteins
11.
Mech Dev ; 138 Pt 3: 279-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26459057

ABSTRACT

The Wnt/Planar Cell Polarity (PCP) pathway controls cell morphology and behavior during animal development. Several zebrafish mutants were identified as having perturbed Wnt/PCP signaling. Many of these mutants have defects in craniofacial formation. To better understand the role that Wnt/PCP plays in craniofacial development we set out to identify which of the mutants, known to be associated with the Wnt/PCP pathway, perturb head cartilage formation by disrupting chondrocyte morphology. Here we demonstrate that while vang-like 2 (vangl2), wnt11 and scribbled (scrib) mutants have severe craniofacial morphogenesis defects they do not display the chondrocyte stacking and intercalation problems seen in glypican 4 (gpc4) and wnt5b mutants. The function of Gpc4 or Wnt5b appears to be important for chondrocyte organization, as the neural crest in both mutants is specified, undergoes migration, and differentiates into the same number of cells to compose the craniofacial cartilage elements. We demonstrate that Gpc4 activity is required cell autonomously in the chondrocytes and that the phenotype of single heterozygous mutants is slightly enhanced in embryos double heterozygous for wnt5b and gpc4. This data suggests a novel mechanism for Wnt5b and Gpc4 regulation of chondrocyte behavior that is independent of the core Wnt/PCP molecules and differs from their collaborative action of controlling cell movements during gastrulation.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis/genetics , Glypicans/genetics , Wnt Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Branchial Region/embryology , Branchial Region/metabolism , Cell Count , Cell Movement/genetics , Cell Size , Chondrocytes/cytology , Gastrulation/genetics , Gene Expression Regulation, Developmental , Glypicans/deficiency , Mutation , Neural Crest/embryology , Neural Crest/metabolism , Phenotype , Wnt Proteins/deficiency , Wnt Signaling Pathway/genetics , Wnt-5a Protein , Zebrafish/metabolism , Zebrafish Proteins/deficiency
12.
J Vis Exp ; (94)2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25489842

ABSTRACT

Polycystic kidney disease (PKD) is one of the most common causes of end-stage kidney disease, a devastating disease for which there is no cure. The molecular mechanisms leading to cyst formation in PKD remain somewhat unclear, but many genes are thought to be involved. Wnt5a is a non-canonical glycoprotein that regulates a wide range of developmental processes. Wnt5a works through the planar cell polarity (PCP) pathway that regulates oriented cell division during renal tubular cell elongation. Defects of the PCP pathway have been found to cause kidney cyst formation. Our paper describes a method for developing a zebrafish cystic kidney disease model by knockdown of the wnt5a gene with wnt5a antisense morpholino (MO) oligonucleotides. Tg(wt1b:GFP) transgenic zebrafish were used to visualize kidney structure and kidney cysts following wnt5a knockdown. Two distinct antisense MOs (AUG - and splice-site) were used and both resulted in curly tail down phenotype and cyst formation after wnt5a knockdown. Injection of mouse Wnt5a mRNA, resistant to the MOs due to a difference in primary base pair structure, rescued the abnormal phenotype, demonstrating that the phenotype was not due to "off-target" effects of the morpholino. This work supports the validity of using a zebrafish model to study wnt5a function in the kidney.


Subject(s)
Disease Models, Animal , Polycystic Kidney Diseases/genetics , Wnt Proteins/genetics , Animals , Animals, Genetically Modified , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Oligonucleotides, Antisense/genetics , Polycystic Kidney Diseases/metabolism , Wnt Proteins/deficiency , Wnt Proteins/metabolism , Wnt-5a Protein , Zebrafish
13.
Med Sci (Paris) ; 30(11): 1024-33, 2014 Nov.
Article in French | MEDLINE | ID: mdl-25388585

ABSTRACT

Advances in genomics, bioinformatics and the creation of model organisms have identified many genes associated with polycystic kidney diseases. Historically, these genes were not necessarily associated with ciliopathies, but it appeared that many connections can be made between the cystic kidney disease and function of the primary cilium. Indeed, the proteins encoded by these genes are localized to the cilium itself, to the basal body or are known to regulate the expression and localization of ciliary proteins. The goal of this article is to describe the multiple cellular processes that may lead to the development of renal cysts if they are deregulated. These include changes in proliferation rate, cell polarity or signaling pathways involved in embryonic kidney development. To highlight the role of the primary cilium in cystogenesis, I will discuss several studies investigating the function of ciliary genes and cilia in the kidneys of different model organisms.


Subject(s)
Cilia/physiology , Ciliary Motility Disorders/pathology , Kidney Diseases, Cystic/pathology , Animals , Cell Division , Cell Movement , Cell Polarity , Ciliary Motility Disorders/genetics , Disease Models, Animal , Epithelial Cells/ultrastructure , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Kidney/embryology , Kidney/ultrastructure , Kidney Diseases, Cystic/classification , Kidney Diseases, Cystic/genetics , Mice , Mitosis , Models, Biological , Morphogenesis , Rats , Signal Transduction/physiology , Wnt Proteins/deficiency , Wnt Proteins/genetics , Wnt Proteins/physiology
14.
Nephron Exp Nephrol ; 128(1-2): 80-8, 2014.
Article in English | MEDLINE | ID: mdl-25412793

ABSTRACT

BACKGROUND: Wnt5a is important for the development of various organs and postnatal cellular function. Little is known, however, about the role of Wnt5a in kidney development, although WNT5A mutations were identified in patients with Robinow syndrome, a genetic disease which includes developmental defects in kidneys. Our goal in this study was to determine the role of Wnt5a in kidney development. METHODS: Whole-mount in situ hybridization was used to establish the expression pattern of Wnt5a during kidney development. Zebrafish with wnt5a knockdown and Wnt5a global knockout mice were used to identify kidney phenotypes. RESULTS: In zebrafish, wnt5a knockdown resulted in glomerular cyst formation and dilated renal tubules. In mice, Wnt5a global knockout resulted in pleiotropic, but severe, kidney phenotypes, including agenesis, fused kidney, hydronephrosis and duplex kidney/ureter. CONCLUSIONS: Our data demonstrated the important role of Wnt5a in kidney development. Disrupted Wnt5a resulted in kidney cysts in zebrafish and pleiotropic abnormal kidney development in mice.


Subject(s)
Kidney/embryology , Kidney/physiology , Wnt Proteins/physiology , Zebrafish Proteins/physiology , Animals , Disease Models, Animal , Female , Gene Knockout Techniques , Incidence , Kidney/abnormalities , Kidney Diseases, Cystic/epidemiology , Kidney Diseases, Cystic/etiology , Kidney Diseases, Cystic/physiopathology , Male , Mice , Mice, Knockout , Models, Animal , Wnt Proteins/deficiency , Wnt Proteins/genetics , Wnt-5a Protein , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
15.
Nat Med ; 20(11): 1279-88, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25306233

ABSTRACT

The WNT16 locus is a major determinant of cortical bone thickness and nonvertebral fracture risk in humans. The disability, mortality and costs caused by osteoporosis-induced nonvertebral fractures are enormous. We demonstrate here that Wnt16-deficient mice develop spontaneous fractures as a result of low cortical thickness and high cortical porosity. In contrast, trabecular bone volume is not altered in these mice. Mechanistic studies revealed that WNT16 is osteoblast derived and inhibits human and mouse osteoclastogenesis both directly by acting on osteoclast progenitors and indirectly by increasing expression of osteoprotegerin (Opg) in osteoblasts. The signaling pathway activated by WNT16 in osteoclast progenitors is noncanonical, whereas the pathway activated in osteoblasts is both canonical and noncanonical. Conditional Wnt16 inactivation revealed that osteoblast-lineage cells are the principal source of WNT16, and its targeted deletion in osteoblasts increases fracture susceptibility. Thus, osteoblast-derived WNT16 is a previously unreported key regulator of osteoclastogenesis and fracture susceptibility. These findings open new avenues for the specific prevention or treatment of nonvertebral fractures, a substantial unmet medical need.


Subject(s)
Fractures, Bone/metabolism , Fractures, Bone/prevention & control , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis , Wnt Proteins/genetics , Wnt Proteins/metabolism , Aging/pathology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Lineage/drug effects , Cells, Cultured , Disease Susceptibility , Fractures, Bone/genetics , Fractures, Bone/pathology , Gene Deletion , Gene Expression Regulation/drug effects , Humans , Mice, Inbred C57BL , Organ Size/drug effects , Organ Size/genetics , Osteoblasts/drug effects , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteocytes/drug effects , Osteocytes/metabolism , Osteocytes/pathology , Osteogenesis/drug effects , Osteogenesis/genetics , Osteoprotegerin/metabolism , RANK Ligand/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Skull/pathology , Wnt Proteins/deficiency
16.
Birth Defects Res A Clin Mol Teratol ; 100(10): 772-88, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25257647

ABSTRACT

BACKGROUND: The heritable multifactorial etiology of human nonsyndromic cleft lip with or without cleft palate (CL ± P) is not understood. CL ± P occurs in 15% of neonates in the homozygous A/WySn mouse strain, with a multifactorial genetic etiology, the clf1 and clf2 variant genes. Clf1 acts as a mutant allele of Wnt9b but its coding sequence is normal. An IAP (intracisternal A particle) retrotransposon inserted near the Wnt9b gene is associated with clf1. METHODS: Transcription of noncoding sequence between the IAP and the Wnt9b gene was examined in A/WySn embryos. The levels of Wnt9b transcript and of an "IAP antisense" transcript initiated in the IAP and extending into the noncoding interval were assayed in A/WySn and C57BL/6J whole embryos or heads across embryonic days 8 to 12. Methylation of the 5' LTR of the IAP was examined in E12 A/WySn embryo heads. RESULTS: Mean Wnt9b transcript levels were lower in A/WySn than in C57BL/6J at all ages examined and lower in CL ± P embryos than in their normal littermates. The "IAP antisense" transcript was found in all A/WySn embryos and was highest in CL ± P embryos. The IAP at Wnt9b was generally unmethylated in CL ± P embryos and approximately 50% methylated in normal littermates. CONCLUSION: The clf1 mutation in A/WySn is a "metastable epiallele", in which stochastic deficiency in some individuals of DNA methylation of a retrotransposon uniquely inserted near the Wnt9b gene allows transcriptional activity of the retrotransposon and interference with transcription from Wnt9b. Methylation of metastable epialleles should be investigated in human nonsyndromic CL ± P.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , DNA Methylation/physiology , Embryo, Mammalian/embryology , Wnt Proteins/deficiency , Analysis of Variance , Animals , Base Sequence , Benzothiazoles , DNA Methylation/genetics , Diamines , Embryo, Mammalian/ultrastructure , Genes, Intracisternal A-Particle/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron, Scanning , Molecular Sequence Data , Organic Chemicals , Quinolines , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
17.
Hum Mol Genet ; 23(25): 6807-14, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25082826

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) affect about 1 in 500 births and are a major cause of morbidity in infants. Duplex collecting systems rank among the most common abnormalities of CAKUT, but the molecular basis for this defect is poorly understood. In mice, conditional deletion of Wnt5a in mesoderm results in bilateral duplex kidney and ureter formation. The ureteric buds (UBs) in mutants emerge as doublets from the intermediate mesoderm (IM)-derived nephric duct (ND) without anterior expansion of the glial cell line-derived neurotrophic factor (Gdnf) expression domain in the surrounding mesenchyme. Wnt5a is normally expressed in a graded manner at the posterior end of the IM, but its expression is down-regulated prior to UB outgrowth at E10.5. Furthermore, ablation of Wnt5a in the mesoderm with an inducible Cre at E7.5 results in duplex UBs, whereas ablation at E8.5 yields normal UB outgrowth, demonstrating that Wnt5a functions in IM development well before the formation of the metanephros. In mutants, the posterior ND is duplicated and surrounding Pax2-positive mesenchymal cells persist in the nephric cord, suggesting that disruption of normal ND patterning prompts the formation of duplex ureters and kidneys. Ror2 homozygous mutants, which infrequently yield duplex collecting systems, show a dramatic increase in incidence with the additional deletion of one copy of Wnt5a, implicating this receptor in non-canonical Wnt5a signaling during IM development. This work provides the first evidence of a role of Wnt5a/Ror2 signaling in IM extension and offers new insights into the etiology of CAKUT and possible involvement of Wnt5a/Ror2 mutations.


Subject(s)
Kidney/metabolism , Mesoderm/metabolism , Morphogenesis/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction/genetics , Wnt Proteins/genetics , Animals , Embryo, Mammalian , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Homozygote , Integrases/genetics , Integrases/metabolism , Kidney/growth & development , Kidney/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mesoderm/growth & development , Mesoderm/pathology , Mice , Mice, Transgenic , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Time Factors , Ureter/growth & development , Ureter/metabolism , Ureter/pathology , Wnt Proteins/deficiency , Wnt-5a Protein , Wolffian Ducts/growth & development , Wolffian Ducts/metabolism , Wolffian Ducts/pathology
18.
Dev Biol ; 387(1): 64-72, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24394376

ABSTRACT

Defects of the ventral body wall are prevalent birth anomalies marked by deficiencies in body wall closure, hypoplasia of the abdominal musculature and multiple malformations across a gamut of organs. However, the mechanisms underlying ventral body wall defects remain elusive. Here, we investigated the role of Wnt signaling in ventral body wall development by inactivating Wls or ß-catenin in murine abdominal ectoderm. The loss of Wls in the ventral epithelium, which blocks the secretion of Wnt proteins, resulted in dysgenesis of ventral musculature and genito-urinary tract during embryonic development. Molecular analyses revealed that the dermis and myogenic differentiation in the underlying mesenchymal progenitor cells was perturbed by the loss of ectodermal Wls. The activity of the Wnt-Pitx2 axis was impaired in the ventral mesenchyme of the mutant body wall, which partially accounted for the defects in ventral musculature formation. In contrast, epithelial depletion of ß-catenin or Wnt5a did not resemble the body wall defects in the ectodermal Wls mutant. These findings indicate that ectodermal Wnt signaling instructs the underlying mesodermal specification and abdominal musculature formation during ventral body wall development, adding evidence to the theory that ectoderm-mesenchyme signaling is a potential unifying mechanism for the origin of ventral body wall defects.


Subject(s)
Abdomen/embryology , Intracellular Signaling Peptides and Proteins/physiology , Muscle Development/genetics , Receptors, G-Protein-Coupled/physiology , Wnt Signaling Pathway/genetics , beta Catenin/physiology , Abdomen/growth & development , Animals , Body Patterning/genetics , Cell Differentiation/genetics , Ectoderm/embryology , Ectoderm/growth & development , Ectoderm/metabolism , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mesoderm/embryology , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Transcription Factors/genetics , Urogenital System/embryology , Urogenital System/growth & development , Wnt Proteins/deficiency , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein , beta Catenin/genetics , Homeobox Protein PITX2
19.
Nature ; 503(7476): 392-6, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24141946

ABSTRACT

Many organs with a high cell turnover (for example, skin, intestine and blood) are composed of short-lived cells that require continuous replenishment by somatic stem cells. Ageing results in the inability of these tissues to maintain homeostasis and it is believed that somatic stem-cell ageing is one underlying cause of tissue attrition with age or age-related diseases. Ageing of haematopoietic stem cells (HSCs) is associated with impaired haematopoiesis in the elderly. Despite a large amount of data describing the decline of HSC function on ageing, the molecular mechanisms of this process remain largely unknown, which precludes rational approaches to attenuate stem-cell ageing. Here we report an unexpected shift from canonical to non-canonical Wnt signalling in mice due to elevated expression of Wnt5a in aged HSCs, which causes stem-cell ageing. Wnt5a treatment of young HSCs induces ageing-associated stem-cell apolarity, reduction of regenerative capacity and an ageing-like myeloid-lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuates HSC ageing, whereas stem-cell-intrinsic reduction of Wnt5a expression results in functionally rejuvenated aged HSCs. Our data demonstrate a critical role for stem-cell-intrinsic non-canonical Wnt5a signalling in HSC ageing.


Subject(s)
Cellular Senescence , Hematopoietic Stem Cells/cytology , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Polarity , Female , Haploinsufficiency , Male , Mice , Mice, Inbred C57BL , Phenotype , Rejuvenation , Wnt Proteins/deficiency , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein , cdc42 GTP-Binding Protein/metabolism
20.
Arterioscler Thromb Vasc Biol ; 33(7): 1679-89, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685555

ABSTRACT

OBJECTIVE: Endothelial cells (ECs) can undergo an endothelial-mesenchymal transition with tissue fibrosis. Wnt- and Msx2-regulated signals participate in arteriosclerotic fibrosis and calcification. We studied the impact of Wnt7, Msx2, and Dkk1, a Wnt7 antagonist, on endothelial-mesenchymal transition in primary aortic ECs. APPROACH AND RESULTS: Transduction of aortic ECs with vectors expressing Dkk1 suppressed EC differentiation and induced a mineralizing myofibroblast phenotype. Dkk1 suppressed claudin 5, PECAM, cadherin 5 (Cdh5), Tie1, and Tie2. Dkk1 converted the cuboidal cell monolayer into a spindle-shaped multilayer and inhibited EC cord formation. Myofibroblast and osteogenic markers, SM22, type I collagen, Osx, Runx2, and alkaline phosphatase, were upregulated by Dkk1 via activin-like kinase/Smad pathways. Dkk1 increased fibrotic mineralization of aortic ECs cultured under osteogenic conditions--the opposite of mesenchymal cell responses. Msx2 and Wnt7b maintained morphology and upregulated markers of differentiated ECs. Deleting EC Wnt7b with the Cdh5-Cre transgene in Wnt7b(fl/fl);LDLR(-/-) mice upregulated aortic osteogenic genes (Osx, Sox9, Runx2, and Msx2) and nuclear phospho-Smad1/5, and increased collagen and calcium accumulation. CONCLUSIONS: Dkk1 enhances endothelial-mesenchymal transition in aortic ECs, whereas Wnt7b and Msx2 signals preserve EC phenotype. EC responses to Dkk1, Wnt7b, and Msx2 are the opposite of mesenchymal responses, coupling EC phenotypic stability with osteofibrogenic predilection during arteriosclerosis.


Subject(s)
Aorta/metabolism , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myofibroblasts/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Biomarkers/metabolism , Cadherins/genetics , Cadherins/metabolism , Cattle , Cell Differentiation , Cell Shape , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Fibrosis , Gene Expression Regulation , Homeodomain Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Myofibroblasts/pathology , Neovascularization, Physiologic , Ossification, Heterotopic/metabolism , Phenotype , Receptors, LDL/genetics , Receptors, LDL/metabolism , Transduction, Genetic , Transfection , Wnt Proteins/deficiency , Wnt Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL