Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.297
Filter
Add more filters

Publication year range
1.
Cell ; 186(2): 428-445.e27, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36626902

ABSTRACT

O-GlcNAc is a dynamic post-translational modification (PTM) that regulates protein functions. In studying the regulatory roles of O-GlcNAc, a major roadblock is the inability to change O-GlcNAcylation on a single protein at a time. Herein, we developed a dual RNA-aptamer-based approach that simultaneously targeted O-GlcNAc transferase (OGT) and ß-catenin, the key transcription factor of the Wnt signaling pathway, to selectively increase O-GlcNAcylation of the latter without affecting other OGT substrates. Using the OGT/ß-catenin dual-specificity aptamers, we found that O-GlcNAcylation of ß-catenin stabilizes the protein by inhibiting its interaction with ß-TrCP. O-GlcNAc also increases ß-catenin's interaction with EZH2, recruits EZH2 to promoters, and dramatically alters the transcriptome. Further, by coupling riboswitches or an inducible expression system to aptamers, we enabled inducible regulation of protein-specific O-GlcNAcylation. Together, our findings demonstrate the efficacy and versatility of dual-specificity aptamers for regulating O-GlcNAcylation on individual proteins.


Subject(s)
Aptamers, Nucleotide , beta Catenin/metabolism , Protein Processing, Post-Translational , Wnt Signaling Pathway , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Acetylglucosamine/metabolism
2.
Cell ; 184(1): 194-206.e14, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33357447

ABSTRACT

Wnts are evolutionarily conserved ligands that signal at short range to regulate morphogenesis, cell fate, and stem cell renewal. The first and essential steps in Wnt secretion are their O-palmitoleation and subsequent loading onto the dedicated transporter Wntless/evenness interrupted (WLS/Evi). We report the 3.2 Å resolution cryogenic electron microscopy (cryo-EM) structure of palmitoleated human WNT8A in complex with WLS. This is accompanied by biochemical experiments to probe the physiological implications of the observed association. The WLS membrane domain has close structural homology to G protein-coupled receptors (GPCRs). A Wnt hairpin inserts into a conserved hydrophobic cavity in the GPCR-like domain, and the palmitoleate protrudes between two helices into the bilayer. A conformational switch of highly conserved residues on a separate Wnt hairpin might contribute to its transfer to receiving cells. This work provides molecular-level insights into a central mechanism in animal body plan development and stem cell biology.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/metabolism , Amino Acid Sequence , Animals , Disulfides/metabolism , Glycosylation , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Signaling Peptides and Proteins/isolation & purification , Models, Molecular , Protein Binding , Protein Domains , Protein Structure, Secondary , Protein Transport , Receptors, G-Protein-Coupled/isolation & purification , Receptors, G-Protein-Coupled/ultrastructure , Structural Homology, Protein , Structure-Activity Relationship , Wnt Proteins/chemistry , Wnt Proteins/isolation & purification , Wnt Proteins/ultrastructure
3.
Annu Rev Cell Dev Biol ; 37: 415-440, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34288710

ABSTRACT

The majority of animal phyla have species that can regenerate. Comparing regeneration across animals can reconstruct the molecular and cellular evolutionary history of this process. Recent studies have revealed some similarity in regeneration mechanisms, but rigorous comparative methods are needed to assess whether these resemblances are ancestral pathways (homology) or are the result of convergent evolution (homoplasy). This review aims to provide a framework for comparing regeneration across animals, focusing on gene regulatory networks (GRNs), which are substrates for assessing process homology. The homology of the wound-induced activation of Wnt signaling and of adult stem cells provides examples of ongoing studies of regeneration that enable comparisons in a GRN framework. Expanding the study of regeneration GRNs in currently studied species and broadening taxonomic sampling for these approaches will identify processes that are unifying principles of regeneration biology across animals. These insights are important both for evolutionary studies of regeneration and for human regenerative medicine.


Subject(s)
Adult Stem Cells , Gene Regulatory Networks , Animals , Gene Regulatory Networks/genetics , Wnt Signaling Pathway/genetics
4.
Cell ; 176(6): 1407-1419.e14, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827680

ABSTRACT

The function of somatic stem cells declines with age. Understanding the molecular underpinnings of this decline is key to counteract age-related disease. Here, we report a dramatic drop in the neural stem cells (NSCs) number in the aging murine brain. We find that this smaller stem cell reservoir is protected from full depletion by an increase in quiescence that makes old NSCs more resistant to regenerate the injured brain. Once activated, however, young and old NSCs show similar proliferation and differentiation capacity. Single-cell transcriptomics of NSCs indicate that aging changes NSCs minimally. In the aging brain, niche-derived inflammatory signals and the Wnt antagonist sFRP5 induce quiescence. Indeed, intervention to neutralize them increases activation of old NSCs during homeostasis and following injury. Our study identifies quiescence as a key feature of old NSCs imposed by the niche and uncovers ways to activate NSCs to repair the aging brain.


Subject(s)
Brain/physiology , Age Factors , Animals , Brain/cytology , Cell Differentiation/physiology , Cell Division/physiology , Cell Proliferation/physiology , Cellular Senescence/physiology , Homeostasis , Male , Mice , Mice, Inbred C57BL , Nerve Regeneration , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , Stem Cell Niche
5.
Cell ; 177(4): 910-924.e22, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30982595

ABSTRACT

The assembly of organized colonies is the earliest manifestation in the derivation or induction of pluripotency in vitro. However, the necessity and origin of this assemblance is unknown. Here, we identify human pluripotent founder cells (hPFCs) that initiate, as well as preserve and establish, pluripotent stem cell (PSC) cultures. PFCs are marked by N-cadherin expression (NCAD+) and reside exclusively at the colony boundary of primate PSCs. As demonstrated by functional analysis, hPFCs harbor the clonogenic capacity of PSC cultures and emerge prior to commitment events or phenotypes associated with pluripotent reprogramming. Comparative single-cell analysis with pre- and post-implantation primate embryos revealed hPFCs share hallmark properties with primitive endoderm (PrE) and can be regulated by non-canonical Wnt signaling. Uniquely informed by primate embryo organization in vivo, our study defines a subset of founder cells critical to the establishment pluripotent state.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cell Differentiation , Cell Lineage , Embryonic Development , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Gene Expression/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Single-Cell Analysis , Wnt Signaling Pathway
6.
Cell ; 174(4): 856-869.e17, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30096312

ABSTRACT

Recent sequencing analyses have shed light on heterogeneous patterns of genomic aberrations in human gastric cancers (GCs). To explore how individual genetic events translate into cancer phenotypes, we established a biological library consisting of genetically engineered gastric organoids carrying various GC mutations and 37 patient-derived organoid lines, including rare genomically stable GCs. Phenotype analyses of GC organoids revealed divergent genetic and epigenetic routes to gain Wnt and R-spondin niche independency. An unbiased phenotype-based genetic screening identified a significant association between CDH1/TP53 compound mutations and the R-spondin independency that was functionally validated by CRISPR-based knockout. Xenografting of GC organoids further established the feasibility of Wnt-targeting therapy for Wnt-dependent GCs. Our results collectively demonstrate that multifaceted genetic abnormalities render human GCs independent of the stem cell niche and highlight the validity of the genotype-phenotype screening strategy in gaining deeper understanding of human cancers.


Subject(s)
Adenocarcinoma/pathology , Organoids/pathology , Stomach Neoplasms/pathology , Stomach/pathology , Thrombospondins/metabolism , Wnt Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Antigens, CD/genetics , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cadherins/genetics , Carcinogenesis , Cell Proliferation , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Organoids/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Thrombospondins/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Wnt Proteins/genetics , Xenograft Model Antitumor Assays
7.
Cell ; 174(4): 870-883.e17, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30057120

ABSTRACT

The mitochondrial unfolded protein response (UPRmt) can be triggered in a cell-non-autonomous fashion across multiple tissues in response to mitochondrial dysfunction. The ability to communicate information about the presence of mitochondrial stress enables a global response that can ultimately better protect an organism from local mitochondrial challenges. We find that animals use retromer-dependent Wnt signaling to propagate mitochondrial stress signals from the nervous system to peripheral tissues. Specifically, the polyQ40-triggered activation of mitochondrial stress or reduction of cco-1 (complex IV subunit) in neurons of C. elegans results in the Wnt-dependent induction of cell-non-autonomous UPRmt in peripheral cells. Loss-of-function mutations of retromer complex components that are responsible for recycling the Wnt secretion-factor/MIG-14 prevent Wnt secretion and thereby suppress cell-non-autonomous UPRmt. Neuronal expression of the Wnt ligand/EGL-20 is sufficient to induce cell-non-autonomous UPRmt in a retromer complex-, Wnt signaling-, and serotonin-dependent manner, clearly implicating Wnt signaling as a strong candidate for the "mitokine" signal.


Subject(s)
Animals, Genetically Modified/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Mitochondria/metabolism , Polyubiquitin/metabolism , Unfolded Protein Response/physiology , Wnt Proteins/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Intracellular Signaling Peptides and Proteins , Mitochondria/genetics , Neurons/cytology , Neurons/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Wnt Proteins/genetics
8.
Cell ; 175(4): 1105-1118.e17, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30343898

ABSTRACT

Neural induction in vertebrates generates a CNS that extends the rostral-caudal length of the body. The prevailing view is that neural cells are initially induced with anterior (forebrain) identity; caudalizing signals then convert a proportion to posterior fates (spinal cord). To test this model, we used chromatin accessibility to define how cells adopt region-specific neural fates. Together with genetic and biochemical perturbations, this identified a developmental time window in which genome-wide chromatin-remodeling events preconfigure epiblast cells for neural induction. Contrary to the established model, this revealed that cells commit to a regional identity before acquiring neural identity. This "primary regionalization" allocates cells to anterior or posterior regions of the nervous system, explaining how cranial and spinal neurons are generated at appropriate axial positions. These findings prompt a revision to models of neural induction and support the proposed dual evolutionary origin of the vertebrate CNS.


Subject(s)
Chromatin Assembly and Disassembly , Embryonic Induction , Neurogenesis , Animals , Cell Line , Cells, Cultured , Chick Embryo , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Spinal Cord/cytology , Spinal Cord/growth & development , Spinal Cord/metabolism
9.
Cell ; 170(6): 1149-1163.e12, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28886383

ABSTRACT

The diversity of mesenchymal cell types in the lung that influence epithelial homeostasis and regeneration is poorly defined. We used genetic lineage tracing, single-cell RNA sequencing, and organoid culture approaches to show that Lgr5 and Lgr6, well-known markers of stem cells in epithelial tissues, are markers of mesenchymal cells in the adult lung. Lgr6+ cells comprise a subpopulation of smooth muscle cells surrounding airway epithelia and promote airway differentiation of epithelial progenitors via Wnt-Fgf10 cooperation. Genetic ablation of Lgr6+ cells impairs airway injury repair in vivo. Distinct Lgr5+ cells are located in alveolar compartments and are sufficient to promote alveolar differentiation of epithelial progenitors through Wnt activation. Modulating Wnt activity altered differentiation outcomes specified by mesenchymal cells. This identification of region- and lineage-specific crosstalk between epithelium and their neighboring mesenchymal partners provides new understanding of how different cell types are maintained in the adult lung.


Subject(s)
Lung/cytology , Mesoderm/cytology , Animals , Homeostasis , Lung/physiology , Mice , Organoids/cytology , Pulmonary Alveoli/cytology , Receptors, G-Protein-Coupled/analysis , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic
10.
Immunity ; 55(12): 2300-2317.e6, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36473468

ABSTRACT

Intestinal stem cell maturation and development coincide with gut microbiota exposure after birth. Here, we investigated how early life microbial exposure, and disruption of this process, impacts the intestinal stem cell niche and development. Single-cell transcriptional analysis revealed impaired stem cell differentiation into Paneth cells and macrophage specification upon antibiotic treatment in early life. Mouse genetic and organoid co-culture experiments demonstrated that a CD206+ subset of intestinal macrophages secreted Wnt ligands, which maintained the mesenchymal niche cells important for Paneth cell differentiation. Antibiotics and reduced numbers of Paneth cells are associated with the deadly infant disease, necrotizing enterocolitis (NEC). We showed that colonization with Lactobacillus or transfer of CD206+ macrophages promoted Paneth cell differentiation and reduced NEC severity. Together, our work defines the gut microbiota-mediated regulation of stem cell niches during early postnatal development.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Mice , Animals , Paneth Cells/physiology , Cell Differentiation/physiology , Macrophages
11.
Mol Cell ; 83(22): 4141-4157.e11, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977121

ABSTRACT

Biomolecular condensates have emerged as a major organizational principle in the cell. However, the formation, maintenance, and dissolution of condensates are still poorly understood. Transcriptional machinery partitions into biomolecular condensates at key cell identity genes to activate these. Here, we report a specific perturbation of WNT-activated ß-catenin condensates that disrupts oncogenic signaling. We use a live-cell condensate imaging method in human cancer cells to discover FOXO and TCF-derived peptides that specifically inhibit ß-catenin condensate formation on DNA, perturb nuclear ß-catenin condensates in cells, and inhibit ß-catenin-driven transcriptional activation and colorectal cancer cell growth. We show that these peptides compete with homotypic intermolecular interactions that normally drive condensate formation. Using this framework, we derive short peptides that specifically perturb condensates and transcriptional activation of YAP and TAZ in the Hippo pathway. We propose a "monomer saturation" model in which short interacting peptides can be used to specifically inhibit condensate-associated transcription in disease.


Subject(s)
Neoplasms , beta Catenin , Humans , beta Catenin/genetics , beta Catenin/metabolism , Signal Transduction , Hippo Signaling Pathway , Peptides/genetics
12.
EMBO J ; 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39349846

ABSTRACT

Polyglutamylation is a reversible posttranslational modification that is catalyzed by enzymes of the tubulin tyrosine ligase-like (TTLL) family. Here, we found that TTLL11 generates a previously unknown type of polyglutamylation that is initiated by the addition of a glutamate residue to the free C-terminal carboxyl group of a substrate protein. TTLL11 efficiently polyglutamylates the Wnt signaling protein Dishevelled 3 (DVL3), thereby changing the interactome of DVL3. Polyglutamylation increases the capacity of DVL3 to get phosphorylated, to undergo phase separation, and to act in the noncanonical Wnt pathway. Both carboxy-terminal polyglutamylation and the resulting reduction in phase separation capacity of DVL3 can be reverted by the deglutamylating enzyme CCP6, demonstrating a causal relationship between TTLL11-mediated polyglutamylation and phase separation. Thus, C-terminal polyglutamylation represents a new type of posttranslational modification, broadening the range of proteins that can be modified by polyglutamylation and providing the first evidence that polyglutamylation can modulate protein phase separation.

13.
Mol Cell ; 79(3): 390-405.e7, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32619402

ABSTRACT

Despite their apparent lack of catalytic activity, pseudokinases are essential signaling molecules. Here, we describe the structural and dynamic properties of pseudokinase domains from the Wnt-binding receptor tyrosine kinases (PTK7, ROR1, ROR2, and RYK), which play important roles in development. We determined structures of all pseudokinase domains in this family and found that they share a conserved inactive conformation in their activation loop that resembles the autoinhibited insulin receptor kinase (IRK). They also have inaccessible ATP-binding pockets, occluded by aromatic residues that mimic a cofactor-bound state. Structural comparisons revealed significant domain plasticity and alternative interactions that substitute for absent conserved motifs. The pseudokinases also showed dynamic properties that were strikingly similar to those of IRK. Despite the inaccessible ATP site, screening identified ATP-competitive type-II inhibitors for ROR1. Our results set the stage for an emerging therapeutic modality of "conformational disruptors" to inhibit or modulate non-catalytic functions of pseudokinases deregulated in disease.


Subject(s)
Cell Adhesion Molecules/chemistry , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Tyrosine Kinase-like Orphan Receptors/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Binding Sites , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Humans , Mice , Models, Molecular , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/chemistry , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/antagonists & inhibitors , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptors, Eph Family/antagonists & inhibitors , Receptors, Eph Family/chemistry , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Spodoptera , Structural Homology, Protein , Substrate Specificity
14.
Am J Hum Genet ; 111(9): 1994-2011, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39168120

ABSTRACT

Zinc and RING finger 3 (ZNRF3) is a negative-feedback regulator of Wnt/ß-catenin signaling, which plays an important role in human brain development. Although somatically frequently mutated in cancer, germline variants in ZNRF3 have not been established as causative for neurodevelopmental disorders (NDDs). We identified 12 individuals with ZNRF3 variants and various phenotypes via GeneMatcher/Decipher and evaluated genotype-phenotype correlation. We performed structural modeling and representative deleterious and control variants were assessed using in vitro transcriptional reporter assays with and without Wnt-ligand Wnt3a and/or Wnt-potentiator R-spondin (RSPO). Eight individuals harbored de novo missense variants and presented with NDD. We found missense variants associated with macrocephalic NDD to cluster in the RING ligase domain. Structural modeling predicted disruption of the ubiquitin ligase function likely compromising Wnt receptor turnover. Accordingly, the functional assays showed enhanced Wnt/ß-catenin signaling for these variants in a dominant negative manner. Contrarily, an individual with microcephalic NDD harbored a missense variant in the RSPO-binding domain predicted to disrupt binding affinity to RSPO and showed attenuated Wnt/ß-catenin signaling in the same assays. Additionally, four individuals harbored de novo truncating or de novo or inherited large in-frame deletion variants with non-NDD phenotypes, including heart, adrenal, or nephrotic problems. In contrast to NDD-associated missense variants, the effects on Wnt/ß-catenin signaling were comparable between the truncating variant and the empty vector and between benign variants and the wild type. In summary, we provide evidence for mirror brain size phenotypes caused by distinct pathomechanisms in Wnt/ß-catenin signaling through protein domain-specific deleterious ZNRF3 germline missense variants.


Subject(s)
Brain , Germ-Line Mutation , Neurodevelopmental Disorders , Phenotype , Ubiquitin-Protein Ligases , Wnt Signaling Pathway , Humans , Wnt Signaling Pathway/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Female , Male , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Brain/metabolism , Brain/pathology , Child , Child, Preschool , beta Catenin/genetics , beta Catenin/metabolism , Adolescent , Mutation, Missense , Genetic Association Studies , Protein Domains
15.
Am J Hum Genet ; 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39419027

ABSTRACT

Microtubule affinity-regulating kinase 2 (MARK2) contributes to establishing neuronal polarity and developing dendritic spines. Although large-scale sequencing studies have associated MARK2 variants with autism spectrum disorder (ASD), the clinical features and variant spectrum in affected individuals with MARK2 variants, early developmental phenotypes in mutant human neurons, and the pathogenic mechanism underlying effects on neuronal development have remained unclear. Here, we report 31 individuals with MARK2 variants and presenting with ASD, other neurodevelopmental disorders, and distinctive facial features. Loss-of-function (LoF) variants predominate (81%) in affected individuals, while computational analysis and in vitro expression assay of missense variants supported the effect of MARK2 loss. Using proband-derived and CRISPR-engineered isogenic induced pluripotent stem cells (iPSCs), we show that MARK2 loss leads to early neuronal developmental and functional deficits, including anomalous polarity and dis-organization in neural rosettes, as well as imbalanced proliferation and differentiation in neural progenitor cells (NPCs). Mark2+/- mice showed abnormal cortical formation and partition and ASD-like behavior. Through the use of RNA sequencing (RNA-seq) and lithium treatment, we link MARK2 loss to downregulation of the WNT/ß-catenin signaling pathway and identify lithium as a potential drug for treating MARK2-associated ASD.

16.
Development ; 151(18)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39250420

ABSTRACT

In vivo and in vitro studies argue that concentration-dependent Wnt signaling regulates mammalian nephron progenitor cell (NPC) programs. Canonical Wnt signaling is regulated through the stabilization of ß-catenin, a transcriptional co-activator when complexed with Lef/Tcf DNA-binding partners. Using the GSK3ß inhibitor CHIR99021 (CHIR) to block GSK3ß-dependent destruction of ß-catenin, we examined dose-dependent responses to ß-catenin in mouse NPCs, using mRNA transduction to modify gene expression. Low CHIR-dependent proliferation of NPCs was blocked on ß-catenin removal, with evidence of NPCs arresting at the G2-M transition. While NPC identity was maintained following ß-catenin removal, mRNA-seq identified low CHIR and ß-catenin dependent genes. High CHIR activated nephrogenesis. Nephrogenic programming was dependent on Lef/Tcf factors and ß-catenin transcriptional activity. Molecular and cellular features of early nephrogenesis were driven in the absence of CHIR by a mutated stabilized form of ß-catenin. Chromatin association studies indicate low and high CHIR response genes are likely direct targets of canonical Wnt transcriptional complexes. Together, these studies provide evidence for concentration-dependent Wnt signaling in the regulation of NPCs and provide new insight into Wnt targets initiating mammalian nephrogenesis.


Subject(s)
Nephrons , Stem Cells , Wnt Signaling Pathway , beta Catenin , Animals , Nephrons/metabolism , Nephrons/cytology , beta Catenin/metabolism , Mice , Stem Cells/metabolism , Stem Cells/cytology , Pyrimidines/pharmacology , Pyridines/pharmacology , Gene Expression Regulation, Developmental , Cell Proliferation , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Organogenesis/genetics , Transcription, Genetic
17.
Development ; 151(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38345319

ABSTRACT

The trunk axial skeleton develops from paraxial mesoderm cells. Our recent study demonstrated that conditional knockout of the stem cell factor Sall4 in mice by TCre caused tail truncation and a disorganized axial skeleton posterior to the lumbar level. Based on this phenotype, we hypothesized that, in addition to the previously reported role of Sall4 in neuromesodermal progenitors, Sall4 is involved in the development of the paraxial mesoderm tissue. Analysis of gene expression and SALL4 binding suggests that Sall4 directly or indirectly regulates genes involved in presomitic mesoderm differentiation, somite formation and somite differentiation. Furthermore, ATAC-seq in TCre; Sall4 mutant posterior trunk mesoderm shows that Sall4 knockout reduces chromatin accessibility. We found that Sall4-dependent open chromatin status drives activation and repression of WNT signaling activators and repressors, respectively, to promote WNT signaling. Moreover, footprinting analysis of ATAC-seq data suggests that Sall4-dependent chromatin accessibility facilitates CTCF binding, which contributes to the repression of neural genes within the mesoderm. This study unveils multiple mechanisms by which Sall4 regulates paraxial mesoderm development by directing activation of mesodermal genes and repression of neural genes.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation, Developmental , Mesoderm , Transcription Factors , Animals , Mice , Cell Differentiation , Chromatin/metabolism , Gene Expression , Mesoderm/metabolism , Somites/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism
18.
Development ; 151(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38358799

ABSTRACT

The Wnt/ß-catenin signaling governs anterior-posterior neural patterning during development. Current human pluripotent stem cell (hPSC) differentiation protocols use a GSK3 inhibitor to activate Wnt signaling to promote posterior neural fate specification. However, GSK3 is a pleiotropic kinase involved in multiple signaling pathways and, as GSK3 inhibition occurs downstream in the signaling cascade, it bypasses potential opportunities for achieving specificity or regulation at the receptor level. Additionally, the specific roles of individual FZD receptors in anterior-posterior patterning are poorly understood. Here, we have characterized the cell surface expression of FZD receptors in neural progenitor cells with different regional identity. Our data reveal unique upregulation of FZD5 expression in anterior neural progenitors, and this expression is downregulated as cells adopt a posterior fate. This spatial regulation of FZD expression constitutes a previously unreported regulatory mechanism that adjusts the levels of ß-catenin signaling along the anterior-posterior axis and possibly contributes to midbrain-hindbrain boundary formation. Stimulation of Wnt/ß-catenin signaling in hPSCs, using a tetravalent antibody that selectively triggers FZD5 and LRP6 clustering, leads to midbrain progenitor differentiation and gives rise to functional dopaminergic neurons in vitro and in vivo.


Subject(s)
Frizzled Receptors , Glycogen Synthase Kinase 3 , beta Catenin , Humans , beta Catenin/metabolism , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Glycogen Synthase Kinase 3/metabolism , Mesencephalon , Nervous System/metabolism , Wnt Signaling Pathway , Animals , Rats
19.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38682276

ABSTRACT

The GPR124/RECK/WNT7 pathway is an essential regulator of CNS angiogenesis and blood-brain barrier (BBB) function. GPR124, a brain endothelial adhesion seven-pass transmembrane protein, associates with RECK, which binds and stabilizes newly synthesized WNT7 that is transferred to frizzled (FZD) to initiate canonical ß-catenin signaling. GPR124 remains enigmatic: although its extracellular domain (ECD) is essential, the poorly conserved intracellular domain (ICD) appears to be variably required in mammals versus zebrafish, potentially via adaptor protein bridging of GPR124 and FZD ICDs. GPR124 ICD deletion impairs zebrafish angiogenesis, but paradoxically retains WNT7 signaling upon mammalian transfection. We thus investigated GPR124 ICD function using the mouse deletion mutant Gpr124ΔC. Despite inefficiently expressed GPR124ΔC protein, Gpr124ΔC/ΔC mice could be born with normal cerebral cortex angiogenesis, in comparison with Gpr124-/- embryonic lethality, forebrain avascularity and hemorrhage. Gpr124ΔC/ΔC vascular phenotypes were restricted to sporadic ganglionic eminence angiogenic defects, attributable to impaired GPR124ΔC protein expression. Furthermore, Gpr124ΔC and the recombinant GPR124 ECD rescued WNT7 signaling in culture upon brain endothelial Gpr124 knockdown. Thus, in mice, GPR124-regulated CNS forebrain angiogenesis and BBB function are exerted by ICD-independent functionality, extending the signaling mechanisms used by adhesion seven-pass transmembrane receptors.


Subject(s)
Blood-Brain Barrier , Brain , Neovascularization, Physiologic , Receptors, G-Protein-Coupled , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/embryology , Neovascularization, Physiologic/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Mice , Brain/metabolism , Brain/embryology , Protein Domains , Mice, Knockout , Signal Transduction , Wnt Proteins/metabolism , Wnt Proteins/genetics , Humans , Endothelial Cells/metabolism , Angiogenesis , GPI-Linked Proteins
20.
Proc Natl Acad Sci U S A ; 121(37): e2406854121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39231208

ABSTRACT

Alzheimer's disease (AD) is a prevalent neurodegenerative disease characterized by cognitive decline and learning/memory impairment associated with neuronal cell loss. Estrogen-related receptor α (ERRα) and ERRγ, which are highly expressed in the brain, have emerged as potential AD regulators, with unelucidated underlying mechanisms. Here, we identified genome-wide binding sites for ERRα and ERRγ in human neuronal cells. They commonly target a subset of genes associated with neurodegenerative diseases, including AD. Notably, Dickkopf-1 (DKK1), a Wnt signaling pathway antagonist, was transcriptionally repressed by both ERRα and ERRγ in human neuronal cells and brain. ERRα and ERRγ repress RNA polymerase II (RNAP II) accessibility at the DKK1 promoter by modulating a specific active histone modification, histone H3 lysine acetylation (H3K9ac), with the potential contribution of their corepressor. This transcriptional repression maintains Wnt signaling activity, preventing tau phosphorylation and promoting a healthy neuronal state in the context of AD.


Subject(s)
Alzheimer Disease , ERRalpha Estrogen-Related Receptor , Intercellular Signaling Peptides and Proteins , Receptors, Estrogen , Animals , Humans , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Brain/metabolism , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Neurons/metabolism , Phosphorylation , Promoter Regions, Genetic , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , tau Proteins/metabolism , tau Proteins/genetics , Wnt Signaling Pathway/genetics
SELECTION OF CITATIONS
SEARCH DETAIL