Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Nature ; 588(7836): 151-156, 2020 12.
Article in English | MEDLINE | ID: mdl-33149305

ABSTRACT

Lymphotoxin ß-receptor (LTßR) signalling promotes lymphoid neogenesis and the development of tertiary lymphoid structures1,2, which are associated with severe chronic inflammatory diseases that span several organ systems3-6. How LTßR signalling drives chronic tissue damage particularly in the lung, the mechanism(s) that regulate this process, and whether LTßR blockade might be of therapeutic value have remained unclear. Here we demonstrate increased expression of LTßR ligands in adaptive and innate immune cells, enhanced non-canonical NF-κB signalling, and enriched LTßR target gene expression in lung epithelial cells from patients with smoking-associated chronic obstructive pulmonary disease (COPD) and from mice chronically exposed to cigarette smoke. Therapeutic inhibition of LTßR signalling in young and aged mice disrupted smoking-related inducible bronchus-associated lymphoid tissue, induced regeneration of lung tissue, and reverted airway fibrosis and systemic muscle wasting. Mechanistically, blockade of LTßR signalling dampened epithelial non-canonical activation of NF-κB, reduced TGFß signalling in airways, and induced regeneration by preventing epithelial cell death and activating WNT/ß-catenin signalling in alveolar epithelial progenitor cells. These findings suggest that inhibition of LTßR signalling represents a viable therapeutic option that combines prevention of tertiary lymphoid structures1 and inhibition of apoptosis with tissue-regenerative strategies.


Subject(s)
Lung/drug effects , Lung/physiology , Lymphotoxin beta Receptor/antagonists & inhibitors , Regeneration/drug effects , Signal Transduction/drug effects , Wnt Proteins/agonists , Adaptive Immunity , Aging/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Apoptosis/drug effects , Emphysema/metabolism , Female , Humans , Immunity, Innate , Lung/metabolism , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Smoke/adverse effects , Stem Cells/drug effects , Stem Cells/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
2.
J Biol Chem ; 298(2): 101586, 2022 02.
Article in English | MEDLINE | ID: mdl-35032551

ABSTRACT

Signaling by bone morphogenetic proteins (BMPs) plays pivotal roles in embryogenesis, adult tissue homeostasis, and disease. Recent studies revealed that the well-established WNT agonist R-spondin 2 (RSPO2) is also a BMP receptor (BMP receptor type 1A) antagonist, with roles in early Xenopus embryogenesis and human acute myeloid leukemia (AML). To uncouple the BMP antagonist function from the WNT agonist function and to promote development of AML therapeutics, here we identified a 10-mer peptide (RW) derived from the thrombospondin 1 domain of RSPO2, which specifically prevents binding between RSPO2 and BMP receptor type 1A without altering WNT signaling. We also show that a corresponding RW dendrimer (RWd) exhibiting improved half-life relieves inhibition of BMP receptor signaling by RSPO2 in human AML cells, reduces cell growth, and induces differentiation. Moreover, microinjection of RWd in Xenopus embryos ventralizes the dorsoventral embryonic patterning by upregulating BMP signaling without affecting WNT signaling. Our study corroborates the function of RSPO2 as a BMP receptor antagonist and provides a proof of concept for pharmacologically uncoupling BMP antagonist from WNT agonist functions of RSPO2 using the inhibitor peptide RWd with enhanced target selectivity and limited side effects.


Subject(s)
Bone Morphogenetic Protein Receptors , Dendrimers , Leukemia, Myeloid, Acute , Wnt Proteins , Adult , Animals , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Proteins , Dendrimers/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Peptide Fragments , Proteins/pharmacology , Wnt Proteins/agonists , Wnt Signaling Pathway , Xenopus laevis
3.
Nature ; 545(7653): 234-237, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28467818

ABSTRACT

Wnt proteins modulate cell proliferation and differentiation and the self-renewal of stem cells by inducing ß-catenin-dependent signalling through the Wnt receptor frizzled (FZD) and the co-receptors LRP5 and LRP6 to regulate cell fate decisions and the growth and repair of several tissues. The 19 mammalian Wnt proteins are cross-reactive with the 10 FZD receptors, and this has complicated the attribution of distinct biological functions to specific FZD and Wnt subtype interactions. Furthermore, Wnt proteins are modified post-translationally by palmitoylation, which is essential for their secretion, function and interaction with FZD receptors. As a result of their acylation, Wnt proteins are very hydrophobic and require detergents for purification, which presents major obstacles to the preparation and application of recombinant Wnt proteins. This hydrophobicity has hindered the determination of the molecular mechanisms of Wnt signalling activation and the functional importance of FZD subtypes, and the use of Wnt proteins as therapeutic agents. Here we develop surrogate Wnt agonists, water-soluble FZD-LRP5/LRP6 heterodimerizers, with FZD5/FZD8-specific and broadly FZD-reactive binding domains. Similar to WNT3A, these Wnt agonists elicit a characteristic ß-catenin signalling response in a FZD-selective fashion, enhance the osteogenic lineage commitment of primary mouse and human mesenchymal stem cells, and support the growth of a broad range of primary human organoid cultures. In addition, the surrogates can be systemically expressed and exhibit Wnt activity in vivo in the mouse liver, regulating metabolic liver zonation and promoting hepatocyte proliferation, resulting in hepatomegaly. These surrogates demonstrate that canonical Wnt signalling can be activated by bi-specific ligands that induce receptor heterodimerization. Furthermore, these easily produced, non-lipidated Wnt surrogate agonists facilitate functional studies of Wnt signalling and the exploration of Wnt agonists for translational applications in regenerative medicine.


Subject(s)
Signal Transduction , Wnt Proteins/agonists , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Frizzled Receptors/metabolism , HEK293 Cells , Hepatocytes/cytology , Hepatomegaly/metabolism , Hepatomegaly/pathology , Humans , Hydrophobic and Hydrophilic Interactions , Intestines/cytology , Ligands , Liver/metabolism , Liver/pathology , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Models, Molecular , Organoids/cytology , Organoids/metabolism , Protein Multimerization , Solubility , Tissue Culture Techniques
4.
Dev Biol ; 448(2): 320-341, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30385275

ABSTRACT

Inhibitors of Apoptosis Protein (IAP) genes participate in processes like apoptosis, proliferation, innate immunity, inflammation, cell motility, differentiation and in malignancies. Here we reveal 25 IAP genes in the tunicate Botryllus schlosseri's genome and their functions in two developmental biology phenomena, a new mode of whole body regeneration (WBR) induced by budectomy, and blastogenesis, the four-staged cycles of botryllid ascidian astogeny. IAP genes that were specifically upregulated during these developmental phenomena were identified, and protein expression patterns of one of these genes, IAP28, were followed. Most of the IAP genes upregulation recorded at blastogenetic stages C/D was in concert with the upregulation at 100 µM H2O2 apoptotic-induced treatment and in parallel to expressions of AIF1, Bax, Mcl1, caspase 2 and two orthologues of caspase 7. Wnt agonist altered the takeover duration along with reduced IAP expressions, and displacement of IAP28+ phagocytes. WBR was initiated solely at blastogenetic stage D, where zooidal absorption was attenuated and regeneration centers were formed either from remains of partially absorbed zooids or from deformed ampullae. Subsequently, bud-bearing zooids developed, in concert with a massive IAP28-dependent phagocytic wave that eliminated the old zooids, then proceeded with the establishment of morphologically normal-looking colonies. IAP4, IAP14 and IAP28 were also involved in WBR, in conjunction with the expression of the pro-survival PI3K-Akt pathway. IAPs function deregulation by Smac mimetics resulted in severe morphological damages, attenuation in bud growth and differentiation, and in destabilization of colonial coordination. Longtime knockdown of IAP functions prior to the budectomy, resulted in colonial death.


Subject(s)
Inhibitor of Apoptosis Proteins/genetics , Regeneration/genetics , Urochordata/genetics , Urochordata/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Gene Expression Regulation, Developmental/drug effects , Hydrogen Peroxide/administration & dosage , Hydrogen Peroxide/toxicity , Inhibitor of Apoptosis Proteins/metabolism , Life Cycle Stages/drug effects , Life Cycle Stages/genetics , Multigene Family , Regeneration/drug effects , Urochordata/drug effects , Urochordata/embryology , Wnt Proteins/agonists , Wnt Proteins/metabolism
5.
Cell Physiol Biochem ; 51(2): 961-978, 2018.
Article in English | MEDLINE | ID: mdl-30466106

ABSTRACT

BACKGROUND/AIMS: Interferon consensus sequence-binding protein 8 (IRF8) belongs to a family of interferon (IFN) regulatory factors that modulates various important physiological processes including carcinogenesis. As reported by others and our group, IRF8 expression is silenced by DNA methylation in both human solid tumors and hematological malignancies. However, the role of IRF8 in lung carcinoma remains elusive. In this study, we determined IRF8 epigenetic regulation, biological functions, and the signaling pathway involved in non-small cell lung cancer (NSCLC). METHODS: IRF8 expression were determined by Q- PCR. MSP and A+T determined promotor methylation. MTS, clonogenic, Transwell assay, Flow cytometry, three-dimensional culture and AO/EB stain verified cell function. In vivo tumorigenesis examed the in vivo effects. By Chip-QPCR, RT-PCR, Western blot and Immunofluorescence staining, the mechanisms were studied. RESULTS: IRF8 was significantly downregulated in lung tumor tissues compared with adjacent non-cancerous tissues. Furthermore, methylation-specific PCR analyses revealed that IRF8 methylation in NSCLC was a common event, and demethylation reagent treatment proved that downregulation of IRF8 was due to its promoter CpG hypermethylation. Clinical data showed that the IRF8 methylation was associated with tumor stage, lymph node metastasis status, patient outcome, and tumor histology. Exogenous expression of IRF8 in the silenced or downregulated lung cancer cell lines A549 and H1299 at least partially restored the sensitivity of lung cancer cells to apoptosis, and arrested cells at the G0/G1 phase. Cell viability, clonogenicity, and cell migration and invasive abilities were strongly inhibited by restored expression of IRF8. A three-dimensional culture system demonstrated that IRF8 changed the cells to a more spherical phenotype. Moreover, ectopic expression of IRF8 enhanced NSCLC chemosensitivity to cisplatin. Furthermore, as verified by Chip-qPCR, immunofluorescence staining, and western blotting, IRF8 bound to the T-cell factor/lymphoid enhancer factor (TCF /LEF) promoter, thus repressing ß-catenin nuclear translocation and its activation. IRF8 significantly disrupted the effects of Wnt agonist, bml284, further suggesting its involvement in the Wnt/ß-catenin pathway. CONCLUSION: IRF8 acted as a tumor suppressor gene through the transcriptional repression of ß-catenin-TCF/LEF in NSCLC. IRF8 methylation may serve as a potential biomarker in NSCLC prognosis.


Subject(s)
Interferon Regulatory Factors/metabolism , Wnt Signaling Pathway , Aged , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Cisplatin/pharmacology , DNA Methylation , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Interferon Regulatory Factors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lymphoid Enhancer-Binding Factor 1/chemistry , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Staging , Promoter Regions, Genetic , Wnt Proteins/agonists , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
6.
J Neurochem ; 139(6): 1175-1191, 2016 12.
Article in English | MEDLINE | ID: mdl-27778356

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder and the most frequent cause of dementia in the aged population. According to the amyloid hypothesis, the amyloid-ß (Aß) peptide plays a key role in the pathogenesis of AD. Aß is generated from the amyloidogenic processing of amyloid precursor protein and can aggregate to form oligomers, which have been described as a major synaptotoxic agent in neurons. Dysfunction of Wnt signaling has been linked to increased Aß formation; however, several other studies have argued against this possibility. Herein, we use multiple experimental approaches to confirm that the inhibition of Wnt signaling promoted the amyloidogenic proteolytic processing of amyloid precursor protein. We also demonstrate that inhibiting Wnt signaling increases the production of the Aß42 peptide, the Aß42 /Aß40 ratio, and the levels of Aß oligomers such as trimers and tetramers. Moreover, we show that activating Wnt signaling reduces the levels of Aß42 and its aggregates, increases Aß40 levels, and reduces the Aß42 /Aß40 ratio. Finally, we show that the protective effects observed in response to activation of the Wnt pathway rely on ß-catenin-dependent transcription, which is demonstrated experimentally via the expression of various 'mutant forms of ß-catenin'. Together, our findings indicate that loss of the Wnt signaling pathway may contribute to the pathogenesis of AD.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Peptide Fragments/biosynthesis , Protein Aggregates/physiology , Wnt Signaling Pathway/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Diterpenes/pharmacology , Humans , Mice , Protein Aggregates/drug effects , Wnt Proteins/agonists , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/biosynthesis , Wnt Signaling Pathway/drug effects , Wnt-5a Protein/agonists , Wnt-5a Protein/antagonists & inhibitors , Wnt-5a Protein/biosynthesis
7.
BMC Biochem ; 17(1): 9, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27207629

ABSTRACT

BACKGROUND: In drug discovery research, cell-based phenotypic screening is an essential method for obtaining potential drug candidates. Revealing the mechanism of action is a key step on the path to drug discovery. However, elucidating the target molecules of hit compounds from phenotypic screening campaigns remains a difficult and troublesome process. Simple and efficient methods for identifying the target molecules are essential. RESULTS: 2-Amino-4-(3,4-(methylenedioxy)benzylamino)-6-(3-methoxyphenyl)pyrimidine (AMBMP) was identified as a senescence inducer from a phenotypic screening campaign. The compound is widely used as a Wnt agonist, although its target molecules remain to be clarified. To identify its target proteins, we compared a series of cellular assay results for the compound with our pathway profiling database. The database comprises the activities of compounds from simple assays of cellular reporter genes and cellular proliferations. In this database, compounds were classified on the basis of statistical analysis of their activities, which corresponded to a mechanism of action by the representative compounds. In addition, the mechanisms of action of the compounds of interest could be predicted using the database. Based on our database analysis, the compound was anticipated to be a tubulin disruptor, which was subsequently confirmed by its inhibitory activity of tubulin polymerization. CONCLUSION: These results demonstrate that tubulin is identified for the first time as a target molecule of the Wnt-activating small molecule and that this might have misled the conclusions of some previous studies. Moreover, the present study also emphasizes that our pathway profiling database is a simple and potent tool for revealing the mechanisms of action of hit compounds obtained from phenotypic screenings and off targets of chemical probes.


Subject(s)
Benzodioxoles/chemistry , Pyrimidines/chemistry , Tubulin/chemistry , Wnt Proteins/agonists , Benzodioxoles/metabolism , Benzodioxoles/pharmacology , Cell Line , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cluster Analysis , Databases, Factual , Genes, Reporter , High-Throughput Screening Assays , Humans , Metabolic Networks and Pathways/drug effects , Microscopy, Fluorescence , Protein Binding , Pyrimidines/metabolism , Pyrimidines/pharmacology , Tubulin/metabolism , Tubulin Modulators/chemistry , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology , Wnt Proteins/metabolism
8.
PLoS Biol ; 10(3): e1001286, 2012.
Article in English | MEDLINE | ID: mdl-22448144

ABSTRACT

Dorsal-ventral specification in the amphibian embryo is controlled by ß-catenin, whose activation in all dorsal cells is dependent on maternal Wnt11. However, it remains unknown whether other maternally secreted factors contribute to ß-catenin activation in the dorsal ectoderm. Here, we show that maternal Xenopus Norrin (xNorrin) promotes anterior neural tissue formation in ventralized embryos. Conversely, when xNorrin function is inhibited, early canonical Wnt signaling in the dorsal ectoderm and the early expression of the zygotic neural inducers Chordin, Noggin, and Xnr3 are severely suppressed, causing the loss of anterior structures. In addition, xNorrin potently inhibits BMP- and Nodal/Activin-related functions through direct binding to the ligands. Moreover, a subset of Norrin mutants identified in humans with Norrie disease retain Wnt activation but show defective inhibition of Nodal/Activin-related signaling in mesoderm induction, suggesting that this disinhibition causes Norrie disease. Thus, xNorrin is an unusual molecule that acts on two major signaling pathways, Wnt and TGF-ß, in opposite ways and is essential for early neuroectoderm specification.


Subject(s)
Neural Plate/embryology , Transforming Growth Factor beta/antagonists & inhibitors , Wnt Signaling Pathway , Xenopus Proteins/metabolism , Xenopus/metabolism , Amino Acid Sequence , Animals , Blindness/congenital , Blindness/genetics , Blindness/metabolism , Blindness/pathology , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Conserved Sequence , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Embryonic Development , Gene Expression Regulation, Developmental , Genetic Diseases, X-Linked , Humans , Ligands , Mesoderm/embryology , Mesoderm/metabolism , Mesoderm/pathology , Molecular Sequence Data , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neural Plate/metabolism , Neural Plate/physiology , Protein Binding , Retinal Degeneration , Spasms, Infantile/genetics , Spasms, Infantile/metabolism , Spasms, Infantile/pathology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Wnt Proteins/agonists , Wnt Proteins/genetics , Wnt Proteins/metabolism , Xenopus/embryology , Xenopus/genetics , Xenopus Proteins/agonists , Xenopus Proteins/genetics
9.
Bioorg Med Chem Lett ; 25(21): 4838-4842, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26152429

ABSTRACT

Both the Wnt/ß-catenin signaling pathway and small GTPases of the ADP-ribosylation factors (ARF) family play important roles in regulating cell development, homeostasis and fate. The previous report of QS11, a small molecule Wnt synergist that binds to ARF GTPase-activating protein 1 (ARFGAP1), suggests a role for ARFGAP1 in the Wnt/ß-catenin pathway. However, direct inhibition of enzymatic activity of ARFGAP1 by QS11 has not been established. Whether ARFGAP1 is the only target that contributes to QS11's Wnt synergy is also not clear. Here we present structure-activity relationship (SAR) studies of QS11 analogs in two assays: direct inhibition of enzymatic activity of purified ARFGAP1 protein and cellular activation of the Wnt/ß-catenin pathway. The results confirm the direct inhibition of ARFGAP1 by QS11, and also suggest the presence of other potential cellular targets of QS11.


Subject(s)
GTPase-Activating Proteins/antagonists & inhibitors , Purines/chemistry , Purines/pharmacology , Wnt Proteins/agonists , Wnt Signaling Pathway/drug effects , Dose-Response Relationship, Drug , GTPase-Activating Proteins/metabolism , Humans , Molecular Structure , Purines/chemical synthesis , Structure-Activity Relationship , beta Catenin/metabolism
10.
Br J Nutr ; 111(9): 1536-48, 2014 May.
Article in English | MEDLINE | ID: mdl-24502851

ABSTRACT

Ketoacids (KA) are known to improve muscle mass among patients with chronic kidney disease (CKD) on a low-protein diet (CKD-LPD), but the mechanism of its preventive effects on muscle atrophy still remains unclear. Since muscle atrophy in CKD may be attributable to the down-regulation of the Wnt7a/Akt/p70S6K pathway and the activation of the ubiquitin-proteasome system (UPS) and the apoptotic signalling pathway, a hypothesis can readily be drawn that KA supplementation improves muscle mass by up-regulating the Wnt7a/Akt/p70S6K pathway and counteracting the activation of the UPS and caspase-3-dependent apoptosis in the muscle of CKD-LPD rats. Rats with 5/6 nephrectomy were randomly divided into three groups, and fed with either 22 % protein (normal-protein diet; NPD), 6 % protein (LPD) or 5 % protein plus 1 % KA for 24 weeks. Sham-operated rats with NPD intake were used as the control. The results demonstrated that KA supplementation improved protein synthesis and increased related mediators such as Wnt7a, phosphorylated Akt and p70S6K in the muscle of CKD-LPD rats. It also inhibited protein degradation, withheld the increase in ubiquitin and its ligases MAFbx (muscle atrophy F-box) and MuRF1 (muscle ring finger-1) as well as attenuated proteasome activity in the muscle of CKD-LPD rats. Moreover, KA supplementation gave rise to a reduction in DNA fragment, cleaved caspase-3 and 14 kDa actin fragment via the down-regulation of the Bax:Bcl-2 ratio in the muscle of CKD-LPD rats. The beneficial effects unveiled herein further consolidate that KA may be a better therapeutic strategy for muscle atrophy in CKD-LPD.


Subject(s)
Dietary Supplements , Disease Models, Animal , Keto Acids/therapeutic use , Muscle, Skeletal/metabolism , Muscular Atrophy/prevention & control , Proto-Oncogene Proteins/agonists , Renal Insufficiency, Chronic/diet therapy , Wnt Proteins/agonists , Animals , Apoptosis , Diet, Protein-Restricted/adverse effects , Down-Regulation , Male , Muscle Proteins/biosynthesis , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Nephrectomy/adverse effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Signal Transduction , Ubiquitination , Up-Regulation , Wnt Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 108(21): 8692-7, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555575

ABSTRACT

Axin proteins are key negative regulators of the canonical Wnt signal transduction pathway. Although Axin2 null mice are viable, we identified an unusual ENU-induced recessive allele of Axin2, canp, that causes midgestation lethality in homozygotes. We show that the Axin2(canp) mutation is a V26D substitution in an invariant N-terminal sequence motif and that the Axin2(canp) protein is more stable than wild type. As predicted for an increased level of a negative regulator, the Axin2(canp) mutation leads to decreased Wnt signaling in most tissues, and this can account for most of the morphological phenotypes of Axin2(canp) mutants. In contrast, there is a paradoxical increase in canonical Wnt activity in the late primitive streak of all Axin2(canp) mutant embryos that is associated with the formation of an ectopic tail in some mutants. Treatment of wild-type embryos with an inhibitor of Tankyrase that stabilizes Axin proteins also causes inhibition of Wnt signaling in anterior regions of the embryo and a gain of Wnt signaling in the primitive streak. The results indicate that although increased stability of Axin2 leads to a loss of canonical Wnt signaling in most tissues, stabilized Axin2 enhances Wnt pathway activity in a specific progenitor population in the late primitive streak.


Subject(s)
Cytoskeletal Proteins/physiology , Signal Transduction/physiology , Wnt Proteins/agonists , Wnt Proteins/antagonists & inhibitors , Animals , Axin Protein , Cytoskeletal Proteins/genetics , Embryo, Mammalian , Mice , Mutation , Organ Specificity , Protein Stability
12.
Cell Chem Biol ; 31(6): 1044-1046, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906109

ABSTRACT

Pharmacological modulation of the Wnt/ß-catenin signaling pathway holds promises for both basic research and therapeutic applications. In this issue of Cell Chemical Biology, Kschonsak et al.1 engineered knotted peptides that promote Wnt signaling by targeting ZNRF3 and serve as pharmacological tools for studying Wnt biology and supporting organoid growth.


Subject(s)
Wnt Signaling Pathway , Wnt Signaling Pathway/drug effects , Humans , Wnt Proteins/metabolism , Wnt Proteins/agonists , Peptides/chemistry , Peptides/pharmacology , Peptides/metabolism , beta Catenin/metabolism , Animals , Receptors, Wnt/metabolism
13.
J Neurosci ; 31(37): 13057-67, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21917789

ABSTRACT

In addition to its role in cellular development and proliferation, there are emerging in vitro data implicating the Wnt/ß-catenin pathway in synaptic plasticity. Yet in vivo studies have not examined whether Wnt activity is required for learning and memory. In the amygdala during fear memory formation, we found that many Wnt-signaling genes were dynamically regulated, with an immediate decrease, followed by an eventual normalization during memory consolidation. This rapid decrease in Wnt mRNA was confirmed with individual quantitative PCR and in situ hybridization. We then manipulated Wnt signaling with a specific peptide antagonist (Dkk-1) or agonist (Wnt1) injected stereotaxically into the adult amygdala during fear learning. We found that neither manipulation had an effect on locomotion, anxiety, fear acquisition, or fear expression. However, both Wnt modulators prevented long-term fear memory consolidation without affecting short-term memory. Dkk-1 and Wnt infusions had destabilizing, but opposite, effects on the requisite ß-catenin/cadherin dynamic interactions that occur during consolidation. These data suggest that dynamic modulation of Wnt/ß-catenin signaling during consolidation is critical for the structural basis of long-term memory formation.


Subject(s)
Amygdala/metabolism , Amygdala/physiology , Conditioning, Classical/physiology , Memory/physiology , Signal Transduction/physiology , Wnt Proteins/physiology , Wnt1 Protein/biosynthesis , Amygdala/drug effects , Animals , Cadherins/metabolism , Conditioning, Classical/drug effects , Fear/drug effects , Fear/physiology , Fear/psychology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Microinjections , Motor Activity/drug effects , Motor Activity/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Wnt Proteins/agonists , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Wnt1 Protein/administration & dosage , Wnt1 Protein/pharmacology , beta Catenin/metabolism
14.
Diabetologia ; 55(1): 255-66, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22016045

ABSTRACT

AIMS/HYPOTHESIS: The wingless-type MMTV integration site (WNT) pathway mediates multiple physiological and pathological processes, such as inflammation, angiogenesis and fibrosis. The aim of this study was to investigate whether canonical WNT signalling plays a role in the pathogenesis of diabetic nephropathy. METHODS: Expression of WNT ligands and frizzled receptors in the canonical WNT pathway in the kidney was compared at the mRNA level using real-time RT-PCR between Akita mice, streptozotocin-induced diabetic rats and db/db mice and their respective non-diabetic controls. Renal function was evaluated by measuring the urine albumin excretion. Human renal proximal tubular epithelial cells were treated with high-glucose medium and 4-hydroxynonenal (HNE). Levels of ß-catenin, connective tissue growth factor and fibronectin were determined by western blot analysis. RESULTS: Some of the WNT ligands and frizzled receptors showed increased mRNA levels in the kidneys of Akita mice, streptozotocin-induced diabetic rats and db/db mice compared with their non-diabetic controls. Renal levels of ß-catenin and WNT proteins were upregulated in these diabetic models. Lowering the blood glucose levels by insulin attenuated the activation of WNT signalling in the kidneys of Akita mice. In cultured human renal proximal tubular epithelial cells, both high glucose and HNE activated WNT signalling. Inhibition of WNT signalling with a monoclonal antibody blocking LDL-receptor-related protein 6 ameliorated renal inflammation and fibrosis and reduced proteinuria in Akita mice. CONCLUSIONS/INTERPRETATION: The WNT pathway is activated in the kidneys of models of both type 1 and 2 diabetes. Dysregulation of the WNT pathway in diabetes represents a new pathogenic mechanism of diabetic nephropathy and renders a new therapeutic target.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney/metabolism , Wnt Signaling Pathway , Animals , Cells, Cultured , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Female , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Humans , Kidney/drug effects , Kidney/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Mice , Mice, Mutant Strains , Molecular Targeted Therapy , Oxidative Stress/drug effects , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , RNA, Messenger/metabolism , Rats , Rats, Inbred BN , Severity of Illness Index , Up-Regulation/drug effects , Wnt Proteins/agonists , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
15.
Proc Natl Acad Sci U S A ; 106(5): 1427-32, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19164755

ABSTRACT

To identify small molecules that can induce beta-cell replication, a large chemical library was screened for proliferation of growth-arrested, reversibly immortalized mouse beta cells by using an automated high-throughput screening platform. A number of structurally diverse, active compounds were identified, including phorbol esters, which likely act through protein kinase C, and a group of thiophene-pyrimidines that stimulate beta-cell proliferation by activating the Wnt signaling pathway. A group of dihydropyridine (DHP) derivatives was also shown to reversibly induce beta-cell replication in vitro by activating L-type calcium channels (LTCCs). Our data suggest that the LTCC agonist 2a affects the expression of genes involved in cell cycle progression and cellular proliferation. Furthermore, treatment of beta cells with both LTCC agonist 2a and the Glp-1 receptor agonist Exendin-4 showed an additive effect on beta-cell replication. The identification of small molecules that induce beta-cell proliferation suggests that it may be possible to reversibly expand other quiescent cells to overcome deficits associated with degenerative and/or autoimmune diseases.


Subject(s)
Calcium Channel Agonists/pharmacology , Cell Proliferation/drug effects , Islets of Langerhans/drug effects , Animals , Calcium Channels, L-Type/drug effects , Cell Line, Transformed , Dihydropyridines/pharmacology , Exenatide , Glucagon-Like Peptide-1 Receptor , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Peptides/pharmacology , Receptors, Glucagon/agonists , Reverse Transcriptase Polymerase Chain Reaction , Venoms/pharmacology , Wnt Proteins/agonists
16.
Glia ; 59(4): 540-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21319221

ABSTRACT

Glycogen synthase kinase 3ß (GSK3ß) is an essential integrating molecule for multiple proliferation and differentiation signals that regulate cell fate. Here, we have examined the effects of inhibiting GSK3ß on the development of oligodendrocytes (OLs) from their oligodendrocyte precursors (OP) in vivo by injection into the lateral ventricle of postnatal mice and ex vivo in organotypic cultures of isolated intact rodent optic nerve. Our results show that a range of GSK3ß inhibitors (ARA-014418, lithium, indirubin, and L803-mt) increase OPs and OLs and promote myelination. Inhibition of GSK3ß stimulates OP proliferation and is prosurvival and antiapoptotic. The effects of GSK3ß inhibition in OPs is via the canonical Wnt signaling pathway by stimulating nuclear translocation of ß-catenin. However, direct comparison of the effects of Wnt3a and GSK3ß inhibition in optic nerves shows that they have opposing actions on OLs, whereby GSK3ß inhibition strikingly increases OL differentiation, whereas Wnt3a inhibits OL differentiation. Notably, GSK3ß inhibition overrides the negative effects of Wnt3a on OLs, indicating novel GSK3ß signaling mechanisms that negatively regulate OL differentiation. We identify that two mechanisms of GSK3ß inhibition are to stimulate cAMP response element binding (CREB) and decrease Notch1 signaling, which positively and negatively regulate OL differentiation and myelination, respectively. A key finding is that GSK3ß inhibition has equivalent effects in the adult and stimulates the regeneration of OLs and remyelination following chemically induced demyelination. This study identifies GSK3ß as a profound negative regulator of OL differentiation that contributes to inefficient regeneration of OLs and myelin repair in demyelination.


Subject(s)
Cell Differentiation/physiology , Glycogen Synthase Kinase 3/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Optic Nerve/metabolism , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Immunohistochemistry , Lithium Chloride/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Oligopeptides/pharmacology , Optic Nerve/drug effects , Organ Culture Techniques , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Wnt Proteins/agonists , Wnt Proteins/metabolism , beta Catenin/metabolism
17.
Dev Growth Differ ; 53(7): 843-56, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21762130

ABSTRACT

WNT signaling activity is involved in the regulation of many cellular functions, including proliferation, migration, cell fate specification, maintenance of pluripotency and induction of tumorigenicity. Here we summarize recent progress towards understanding the regulation of canonical WNT/ß-catenin signaling activity through feedback regulatory loops involving the ligands, agonists and antagonists, the availability of intracellular pools of active ß-catenin and the cross-regulation of the WNT activity by ß-catenin independent pathway. We also review recent findings on the role of WNT/ß-catenin signaling in tissue lineage differentiation during embryogenesis and the maintenance and self renewal of embryo-derived stem cells in vitro.


Subject(s)
Cell Differentiation , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement , Embryo, Mammalian/metabolism , Embryonic Development , Embryonic Stem Cells/cytology , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Germ Layers/cytology , Germ Layers/embryology , Glypicans/metabolism , Heart/embryology , Heart/growth & development , Heparan Sulfate Proteoglycans/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Phenotype , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Wnt Proteins/agonists , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , beta Catenin/genetics , beta Catenin/metabolism
18.
Mol Psychiatry ; 15(3): 272-85, 228, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19621015

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive deterioration of cognitive abilities, accumulation of the amyloid-beta-peptide (Abeta) and synaptic alterations. Treatment with lithium has been shown to provide neuroprotection against several insults, including protection against Abeta neurotoxicity in vitro. Rosiglitazone, a peroxisome proliferator activated receptor-gamma agonist, has been shown to attenuate Abeta-peptide neurotoxic effects, including the inflammatory response of microglia and astrocytes. Both types of drugs activate Wnt signaling, a pathway that has been shown to be related to AD. In this study, a double transgenic mouse model, which coexpresses APPswe and the exon 9 deletion of the presenilin 1 (PSEN1) gene, was used to examine, in vivo, the effect of lithium and rosiglitazone on Abeta neurotoxicity. Mice were tested for spatial memory, and their brain samples were used for histochemical and biochemical analysis. In this study, we report that both drugs significantly reduced (1) spatial memory impairment induced by amyloid burden; (2) Abeta aggregates and Abeta oligomers; and (3) astrocytic and microglia activation. They also prevented changes in presynaptic and postsynaptic marker proteins. Finally, both drugs activate Wnt signaling shown by the increase in beta-catenin and by the inhibition of the glycogen synthase kinase-3beta. We conclude that lithium and rosiglitazone, possibly by the activation of the Wnt signaling pathway, reduce various AD neuropathological markers and may be considered as potential therapeutic agents against the disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/genetics , Lithium Compounds/pharmacology , Memory Disorders/drug therapy , Nerve Degeneration/drug therapy , Presenilin-1/genetics , Thiazolidinediones/pharmacology , Wnt Proteins/agonists , Alzheimer Disease/genetics , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Brain/cytology , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Inflammation/drug therapy , Lithium Compounds/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Microglia/drug effects , Nerve Tissue Proteins/drug effects , Rosiglitazone , Signal Transduction/drug effects , Thiazolidinediones/therapeutic use
19.
Biol Open ; 10(11)2021 11 15.
Article in English | MEDLINE | ID: mdl-34643229

ABSTRACT

Bovine embryonic stem cells (ESC) have features associated with the primed pluripotent state including low expression of one of the core pluripotency transcription factors, NANOG. It has been reported that NANOG expression can be upregulated in porcine ESC by treatment with activin A and the WNT agonist CHIR99021. Accordingly, it was tested whether expression of NANOG and another pluripotency factor SOX2 could be stimulated by activin A and the WNT agonist CHIR99021. Immunoreactive NANOG and SOX2 were analyzed for bovine ESC lines derived under conditions in which activin A and CHIR99021 were added singly or in combination. Activin A enhanced NANOG expression but also reduced SOX2 expression. CHIR99021 depressed expression of both NANOG and SOX2. In a second experiment, activin A enhanced blastocyst development while CHIR99021 treatment impaired blastocyst formation and reduced number of blastomeres. Activin A treatment decreased blastomeres in the blastocyst that were positive for either NANOG or SOX2 but increased those that were CDX2+ and that were GATA6+ outside the inner cell mass. CHIR99021 reduced SOX2+ and NANOG+ blastomeres without affecting the number or percent of blastomeres that were CDX2+ and GATA6+. Results indicate activation of activin A signaling stimulates NANOG expression during self-renewal of bovine ESC but suppresses cells expressing pluripotency markers in the blastocyst and increases cells expressing CDX2. Actions of activin A to promote blastocyst development may involve its role in promoting trophectoderm formation. Furthermore, results demonstrate the negative role of canonical WNT signaling in cattle for pluripotency marker expression in ESC and in formation of the inner cell mass and epiblast during embryonic development. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Activins/metabolism , Blastocyst/metabolism , Embryonic Stem Cells/metabolism , Nanog Homeobox Protein/metabolism , SOXB1 Transcription Factors/metabolism , Wnt Proteins/agonists , Animals , Cattle , Cell Line , Embryonic Development/genetics , Germ Layers , Pyridines/metabolism , Pyrimidines/metabolism , Swine , Wnt Signaling Pathway/genetics
20.
Nat Commun ; 12(1): 3247, 2021 05 31.
Article in English | MEDLINE | ID: mdl-34059688

ABSTRACT

The Wnt signaling pathway is intricately connected with bone mass regulation in humans and rodent models. We designed an antibody-based platform that generates potent and selective Wnt mimetics. Using this platform, we engineer bi-specific Wnt mimetics that target Frizzled and low-density lipoprotein receptor-related proteins and evaluate their effects on bone accrual in murine models. These synthetic Wnt agonists induce rapid and robust bone building effects, and correct bone mass deficiency and bone defects in various disease models, including osteoporosis, aging, and long bone fracture. Furthermore, when these Wnt agonists are combined with antiresorptive bisphosphonates or anti-sclerostin antibody therapies, additional bone accrual/maintenance effects are observed compared to monotherapy, which could benefit individuals with severe and/or acute bone-building deficiencies. Our data support the continued development of Wnt mimetics for the treatment of diseases of low bone mineral density, including osteoporosis.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Resorption/drug therapy , Femoral Fractures/drug therapy , Osteoporosis, Postmenopausal/drug therapy , Wnt Proteins/agonists , Aged , Aging/physiology , Animals , Bone Density/drug effects , Bone Density/physiology , Bone Density Conservation Agents/therapeutic use , Bone Resorption/physiopathology , Diphosphonates/pharmacology , Diphosphonates/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination/methods , Female , Femoral Fractures/pathology , Femur/drug effects , Femur/injuries , Femur/pathology , Humans , Mice , Osteoporosis, Postmenopausal/physiopathology , Wnt Signaling Pathway/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL