Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
Add more filters

Publication year range
1.
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
2.
Mol Cell ; 83(10): 1677-1692.e8, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37207626

ABSTRACT

PERIOD (PER) and Casein Kinase 1δ regulate circadian rhythms through a phosphoswitch that controls PER stability and repressive activity in the molecular clock. CK1δ phosphorylation of the familial advanced sleep phase (FASP) serine cluster embedded within the Casein Kinase 1 binding domain (CK1BD) of mammalian PER1/2 inhibits its activity on phosphodegrons to stabilize PER and extend circadian period. Here, we show that the phosphorylated FASP region (pFASP) of PER2 directly interacts with and inhibits CK1δ. Co-crystal structures in conjunction with molecular dynamics simulations reveal how pFASP phosphoserines dock into conserved anion binding sites near the active site of CK1δ. Limiting phosphorylation of the FASP serine cluster reduces product inhibition, decreasing PER2 stability and shortening circadian period in human cells. We found that Drosophila PER also regulates CK1δ via feedback inhibition through the phosphorylated PER-Short domain, revealing a conserved mechanism by which PER phosphorylation near the CK1BD regulates CK1 kinase activity.


Subject(s)
Circadian Clocks , Period Circadian Proteins , Animals , Humans , Phosphorylation , Feedback , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Casein Kinase I/genetics , Casein Kinase I/metabolism , Circadian Rhythm/genetics , Drosophila/metabolism , Serine/metabolism , Mammals/metabolism
3.
Mol Cell ; 81(6): 1133-1146, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33545069

ABSTRACT

In our 24/7 well-lit world, it's easy to skip or delay sleep to work, study, and play. However, our circadian rhythms are not easily fooled; the consequences of jet lag and shift work are many and severe, including metabolic, mood, and malignant disorders. The internal clock that keeps track of time has at its heart the reversible phosphorylation of the PERIOD proteins, regulated by isoforms of casein kinase 1 (CK1). In-depth biochemical, genetic, and structural studies of these kinases, their mutants, and their splice variants have combined over the past several years to provide a robust understanding of how the core clock is regulated by a phosphoswitch whereby phosphorylation of a stabilizing site on PER blocks phosphorylation of a distant phosphodegron. The recent structure of a circadian mutant form of CK1 implicates an internal activation loop switch that regulates this phosphoswitch and points to new approaches to regulation of the clock.


Subject(s)
Casein Kinase I/metabolism , Circadian Clocks/physiology , Circadian Rhythm/physiology , Period Circadian Proteins/metabolism , Animals , Casein Kinase I/genetics , Humans , Period Circadian Proteins/genetics , Phosphorylation/physiology
4.
J Biol Chem ; 300(6): 107391, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38777144

ABSTRACT

The duration of the transcription-repression cycles that give rise to mammalian circadian rhythms is largely determined by the stability of the PERIOD (PER) protein, the rate-limiting components of the molecular clock. The degradation of PERs is tightly regulated by multisite phosphorylation by casein kinase 1 (CK1δ/ε). In this phosphoswitch, phosphorylation of a PER2 degron [degron 2 (D2)] causes degradation, while phosphorylation of the PER2 familial advanced sleep phase (FASP) domain blocks CK1 activity on the degron, stabilizing PER2. However, this model and many other studies of PER2 degradation do not include the second degron of PER2 that is conserved in PER1, termed degron 1 (D1). We examined how these two degrons contribute to PER2 stability, affect the balance of the phosphoswitch, and how they are differentiated by CK1. Using PER2-luciferase fusions and real-time luminometry, we investigated the contribution of both D2 and of CK1-PER2 binding. We find that D1, like D2, is a substrate of CK1 but that D1 plays only a 'backup' role in PER2 degradation. Notably, CK1 bound to a PER1:PER2 dimer protein can phosphorylate PER1 D1 in trans. This scaffolded phosphorylation provides additional levels of control to PER stability and circadian rhythms.


Subject(s)
Period Circadian Proteins , Protein Stability , Period Circadian Proteins/metabolism , Period Circadian Proteins/genetics , Phosphorylation , Humans , HEK293 Cells , Proteolysis , Animals , Casein Kinase I/metabolism , Casein Kinase I/genetics , Circadian Rhythm , Mice , Degrons
5.
Mol Cell ; 66(4): 437-438, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28525736

ABSTRACT

Circadian clocks signal and adapt to an ever-changing world by juggling a panoply of transcriptional and post-translational modifications. In this issue of Molecular Cell, Gustafson et al. (2017) report an additional requirement for accurate timekeeping, a cis/trans conformational flicker in the transcriptional activation domain of the core clock protein BMAL1.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Clocks , CLOCK Proteins/genetics , Circadian Rhythm , Protein Processing, Post-Translational
6.
N Engl J Med ; 385(14): 1292-1301, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34587386

ABSTRACT

BACKGROUND: Structural birth defects occur in approximately 3% of live births; most such defects lack defined genetic or environmental causes. Despite advances in surgical approaches, pharmacologic prevention remains largely out of reach. METHODS: We queried worldwide databases of 20,248 families that included children with neurodevelopmental disorders and that were enriched for parental consanguinity. Approximately one third of affected children in these families presented with structural birth defects or microcephaly. We performed exome or genome sequencing of samples obtained from the children, their parents, or both to identify genes with biallelic pathogenic or likely pathogenic mutations present in more than one family. After identifying disease-causing variants, we generated two mouse models, each with a pathogenic variant "knocked in," to study mechanisms and test candidate treatments. We administered a small-molecule Wnt agonist to pregnant animals and assessed their offspring. RESULTS: We identified homozygous mutations in WLS, which encodes the Wnt ligand secretion mediator (also known as Wntless or WLS) in 10 affected persons from 5 unrelated families. (The Wnt ligand secretion mediator is essential for the secretion of all Wnt proteins.) Patients had multiorgan defects, including microcephaly and facial dysmorphism as well as foot syndactyly, renal agenesis, alopecia, iris coloboma, and heart defects. The mutations affected WLS protein stability and Wnt signaling. Knock-in mice showed tissue and cell vulnerability consistent with Wnt-signaling intensity and individual and collective functions of Wnts in embryogenesis. Administration of a pharmacologic Wnt agonist partially restored embryonic development. CONCLUSIONS: Genetic variations affecting a central Wnt regulator caused syndromic structural birth defects. Results from mouse models suggest that what we have named Zaki syndrome is a potentially preventable disorder. (Funded by the National Institutes of Health and others.).


Subject(s)
Abnormalities, Multiple/genetics , Congenital Abnormalities/genetics , Genetic Pleiotropy , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Receptors, G-Protein-Coupled/genetics , Wnt Proteins/metabolism , Animals , Disease Models, Animal , Fibroblasts/metabolism , Gene Knock-In Techniques , Genes, Recessive , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Pedigree , Phenotype , Receptors, G-Protein-Coupled/metabolism , Syndrome , Wnt Signaling Pathway
7.
Cell ; 137(4): 602-4, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19450508

ABSTRACT

Circadian clocks use temperature compensation to keep accurate time over a range of temperatures, thus allowing reliable timekeeping under diverse environmental conditions. Mehra et al. (2009) and Baker et al. (2009) now show that phosphorylation-regulated protein degradation plays a key role in circadian temperature compensation.


Subject(s)
Biological Clocks , Circadian Rhythm , Neurospora crassa/physiology , Casein Kinase II/chemistry , Casein Kinase II/genetics , Casein Kinase II/physiology , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/physiology , Neurospora crassa/enzymology , Phosphorylation , Temperature
8.
J Cell Sci ; 134(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34817055

ABSTRACT

Wnt signaling is essential for normal development and is a therapeutic target in cancer. The enzyme PORCN, or porcupine, is a membrane-bound O-acyltransferase (MBOAT) that is required for the post-translational modification of all Wnts, adding an essential mono-unsaturated palmitoleic acid to a serine on the tip of Wnt hairpin 2. Inherited mutations in PORCN cause focal dermal hypoplasia, and therapeutic inhibition of PORCN slows the growth of Wnt-dependent cancers. Based on homology to mammalian MBOAT proteins, we developed and validated a structural model of human PORCN. The model accommodates palmitoleoyl-CoA and Wnt hairpin 2 in two tunnels in the conserved catalytic core, shedding light on the catalytic mechanism. The model predicts how previously uncharacterized human variants of uncertain significance can alter PORCN function. Drugs including ETC-159, IWP-L6 and LGK-974 dock in the PORCN catalytic site, providing insights into PORCN pharmacologic inhibition. This structural model enhances our mechanistic understanding of PORCN substrate recognition and catalysis, as well as the inhibition of its enzymatic activity, and can facilitate the development of improved inhibitors and the understanding of disease-relevant PORCN mutants. This article has an associated First Person interview with the joint first authors of the paper.


Subject(s)
Focal Dermal Hypoplasia , Pharmaceutical Preparations , Acyltransferases/genetics , Animals , Catalytic Domain , Humans , Membrane Proteins/genetics , Models, Structural
9.
Mol Cell ; 60(1): 77-88, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26431025

ABSTRACT

Period (PER) protein phosphorylation is a critical regulator of circadian period, yet an integrated understanding of the role and interaction between phosphorylation sites that can both increase and decrease PER2 stability remains elusive. Here, we propose a phosphoswitch model, where two competing phosphorylation sites determine whether PER2 has a fast or slow degradation rate. This mathematical model accurately reproduces the three-stage degradation kinetics of endogenous PER2. We predict and demonstrate that the phosphoswitch is intrinsically temperature sensitive, slowing down PER2 degradation as a result of faster reactions at higher temperatures. The phosphoswitch provides a biochemical mechanism for circadian temperature compensation of circadian period. This phosphoswitch additionally explains the phenotype of Familial Advanced Sleep Phase (FASP) and CK1ε(tau) genetic circadian rhythm disorders, metabolic control of PER2 stability, and how drugs that inhibit CK1 alter period. The phosphoswitch provides a general mechanism to integrate diverse stimuli to regulate circadian period.


Subject(s)
Circadian Rhythm , Models, Biological , Period Circadian Proteins/chemistry , Period Circadian Proteins/metabolism , Animals , Cell Line , Mice , NIH 3T3 Cells , Phosphorylation , Protein Stability , Proteolysis , Temperature
10.
Proc Natl Acad Sci U S A ; 117(20): 10888-10896, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32354999

ABSTRACT

Casein kinase 1 (CK1) plays a central role in regulating the period of the circadian clock. In mammals, PER2 protein abundance is regulated by CK1-mediated phosphorylation and proteasomal degradation. On the other hand, recent studies have questioned whether the degradation of the core circadian machinery is a critical step in clock regulation. Prior cell-based studies found that CK1 phosphorylation of PER2 at Ser478 recruits the ubiquitin E3 ligase ß-TrCP, leading to PER2 degradation. Creation of this phosphodegron is regulated by a phosphoswitch that is also implicated in temperature compensation. However, in vivo evidence that this phosphodegron influences circadian period is lacking. Here, we generated and analyzed PER2-Ser478Ala knock-in mice. The mice showed longer circadian period in behavioral analysis. Molecularly, mutant PER2 protein accumulated in both the nucleus and cytoplasm of the mouse liver, while Per2 messenger RNA (mRNA) levels were minimally affected. Nuclear PER1, CRY1, and CRY2 proteins also increased, probably due to stabilization of PER2-containing complexes. In mouse embryonic fibroblasts derived from PER2-Ser478Ala::LUC mice, three-phase decay and temperature compensation of the circadian period was perturbed. These data provide direct in vivo evidence for the importance of phosphorylation-regulated PER2 stability in the circadian clock and validate the phosphoswitch in a mouse model.


Subject(s)
Circadian Clocks/physiology , Mutation , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Animals , Behavior, Animal , Casein Kinase I/metabolism , Cell Nucleus/metabolism , Circadian Rhythm/physiology , Female , Gene Expression Regulation , Liver , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Phosphorylation , RNA, Messenger/metabolism , Transcriptome , Ubiquitin-Protein Ligases/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism
11.
Int J Mol Sci ; 24(5)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36902186

ABSTRACT

There is an increasing urgency in the search for new drugs to target high-grade cancers such as osteosarcomas (OS), as these have limited therapeutic options and poor prognostic outlook. Even though key molecular events leading to tumorigenesis are not well understood, it is widely agreed that OS tumours are Wnt-driven. ETC-159, a PORCN inhibitor that inhibits the extracellular secretion of Wnt, has recently progressed on to clinical trials. In vitro and in vivo murine and chick chorioallantoic membrane xenograft models were established to examine the effect of ETC-159 on OS. Consistent with our hypothesis, we noted that ETC-159 treatment not only resulted in markedly decreased ß-catenin staining in xenografts, but also increased tumour necrosis and a significant reduction in vascularity-a hereby yet undescribed phenotype following ETC-159 treatment. Through further understanding the mechanism of this new window of vulnerability, therapies can be developed to potentiate and maximize the effectiveness of ETC-159, further increasing its clinical utility for the treatment of OS.


Subject(s)
Acyltransferases , Bone Neoplasms , Neovascularization, Pathologic , Osteosarcoma , Wnt Signaling Pathway , Animals , Humans , Mice , Acyltransferases/antagonists & inhibitors , beta Catenin/metabolism , Bone Neoplasms/blood supply , Bone Neoplasms/drug therapy , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation , Membrane Proteins/antagonists & inhibitors , Necrosis , Osteosarcoma/blood supply , Osteosarcoma/drug therapy , Wnt Signaling Pathway/drug effects , Neovascularization, Pathologic/drug therapy
12.
Biochem Soc Trans ; 50(6): 1797-1808, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36484635

ABSTRACT

Wnts are lipid-modified signaling glycoproteins present in all metazoans that play key roles in development and homeostasis. Post-translational modifications of Wnts regulate their function. Wnts have a unique post-translational modification, O-linked palmitoleation, that is absolutely required for their function. This Wnt-specific modification occurs during Wnt biosynthesis in the endoplasmic reticulum (ER), catalyzed by the O-acyltransferase Porcupine (PORCN). Palmitoleation is required for Wnt to bind to its transporter Wntless (WLS/Evi) as well as to its receptor Frizzled (FZD). Recent structural studies have illustrated how PORCN recognizes its substrates, and how drugs inhibit this. The abundance of WLS is tightly regulated by intracellular recycling and ubiquitylation-mediated degradation in the ER. The function of Wnt glycosylation is less well understood, and the sites and types of glycosylation are not largely conserved among different Wnts. In polarized tissues, the type of glycans can determine whether the route of trafficking is apical or basolateral. In addition, pairing of the 24 highly conserved cysteines in Wnts to form disulfide bonds is critical in maintaining proper structure and activities. Extracellularly, the amino terminus of a subset of Wnts can be cleaved by a dedicated glycosylphosphatidylinositol (GPI)-anchored metalloprotease TIKI, resulting in the inactivation of these Wnt proteins. Additionally, NOTUM is a secreted extracellular carboxylesterase that removes the palmitoleate moiety from Wnt, antagonizing its activity. In summary, Wnt signaling activity is controlled at multiple layers by post-translational modifications.


Subject(s)
Protein Processing, Post-Translational , Wnt Proteins , Wnt Proteins/metabolism , Wnt Signaling Pathway , Acyltransferases/metabolism , Endoplasmic Reticulum/metabolism
13.
Cancer Metastasis Rev ; 39(3): 625-645, 2020 09.
Article in English | MEDLINE | ID: mdl-32385713

ABSTRACT

Since the discovery of the first mammalian Wnt proto-oncogene in virus-induced mouse mammary tumors almost four decades ago, Wnt signaling pathway and its involvement in cancers have been extensively investigated. Activation of this evolutionarily conserved pathway promotes cancer development via diverse mechanisms. Cancer is a complex disease and one outstanding conceptual framework for understanding its biology is the "Hallmarks of Cancer". In this review, we focus on the involvement of Wnt signaling in the ten hallmarks of human cancer. These widespread roles of Wnt signaling in human cancers highlight the importance and feasibility of targeting this signaling pathway for cancer treatment.


Subject(s)
Neoplasms/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Proto-Oncogene Mas
14.
RNA ; 25(12): 1696-1713, 2019 12.
Article in English | MEDLINE | ID: mdl-31506381

ABSTRACT

Differential gene isoform expression is a ubiquitous mechanism to enhance proteome diversity and maintain cell homeostasis. Mechanisms such as splicing that drive gene isoform variability are highly dynamic and responsive to changes in cell signaling pathways. Wnt/ß-catenin signaling has profound effects on cell activity and cell fate and is known to modify several splicing events by altering the expression of individual splicing factors. However, a global assessment of how extensively Wnt signaling regulates splicing and other mechanisms that determine mRNA isoform composition in cancer is lacking. We used deep time-resolved RNA-seq in two independent in vivo Wnt-addicted tumor models during treatment with the potent Wnt inhibitor ETC-159 and examined Wnt regulated splicing events and splicing regulators. We found 1025 genes that underwent Wnt regulated variable exon usage leading to isoform expression changes. This was accompanied by extensive Wnt regulated changes in the expression of splicing regulators. Many of these Wnt regulated events were conserved in multiple human cancers, and many were linked to previously defined cancer-associated splicing quantitative trait loci. This suggests that the Wnt regulated splicing events are components of fundamental oncogenic processes. These findings demonstrate the wide-ranging effects of Wnt signaling on the isoform composition of the cell and provides an extensive resource of expression changes of splicing regulators and gene isoforms regulated by Wnt signaling.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/metabolism , Wnt Signaling Pathway , Alternative Splicing , Biomarkers , Cell Line, Tumor , Exons , Gene Expression Profiling , Humans , Neoplasms/pathology , Protein Isoforms , Quantitative Trait Loci , RNA, Messenger/genetics
15.
Proc Natl Acad Sci U S A ; 115(14): E3173-E3181, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29559533

ABSTRACT

Wnts and R-spondins (RSPOs) support intestinal homeostasis by regulating crypt cell proliferation and differentiation. Ex vivo, Wnts secreted by Paneth cells in organoids can regulate the proliferation and differentiation of Lgr5-expressing intestinal stem cells. However, in vivo, Paneth cell and indeed all epithelial Wnt production is completely dispensable, and the cellular source of Wnts and RSPOs that maintain the intestinal stem-cell niche is not known. Here we investigated both the source and the functional role of stromal Wnts and RSPO3 in regulation of intestinal homeostasis. RSPO3 is highly expressed in pericryptal myofibroblasts in the lamina propria and is several orders of magnitude more potent than RSPO1 in stimulating both Wnt/ß-catenin signaling and organoid growth. Stromal Rspo3 ablation ex vivo resulted in markedly decreased organoid growth that was rescued by exogenous RSPO3 protein. Pdgf receptor alpha (PdgfRα) is known to be expressed in pericryptal myofibroblasts. We therefore evaluated if PdgfRα identified the key stromal niche cells. In vivo, Porcn excision in PdgfRα+ cells blocked intestinal crypt formation, demonstrating that Wnt production in the stroma is both necessary and sufficient to support the intestinal stem-cell niche. Mice with Rspo3 excision in the PdgfRα+ cells had decreased intestinal crypt Wnt/ß-catenin signaling and Paneth cell differentiation and were hypersensitive when stressed with dextran sodium sulfate. The data support a model of the intestinal stem-cell niche regulated by both Wnts and RSPO3 supplied predominantly by stromal pericryptal myofibroblasts marked by PdgfRα.


Subject(s)
Epithelial Cells/cytology , Intestines/cytology , Receptor, Platelet-Derived Growth Factor alpha/physiology , Stem Cell Niche/physiology , Stem Cells/cytology , Stromal Cells/cytology , Thrombospondins/metabolism , Wnt1 Protein/metabolism , Acyltransferases/physiology , Animals , Cell Differentiation , Cell Proliferation , Epithelial Cells/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoids/cytology , Organoids/metabolism , Stem Cells/metabolism , Stromal Cells/metabolism , Thrombospondins/genetics , Wnt1 Protein/genetics
16.
Proc Natl Acad Sci U S A ; 115(23): 5986-5991, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29784789

ABSTRACT

Multisite phosphorylation of the PERIOD 2 (PER2) protein is the key step that determines the period of the mammalian circadian clock. Previous studies concluded that an unidentified kinase is required to prime PER2 for subsequent phosphorylation by casein kinase 1 (CK1), an essential clock component that is conserved from algae to humans. These subsequent phosphorylations stabilize PER2, delay its degradation, and lengthen the period of the circadian clock. Here, we perform a comprehensive biochemical and biophysical analysis of mouse PER2 (mPER2) priming phosphorylation and demonstrate, surprisingly, that CK1δ/ε is indeed the priming kinase. We find that both CK1ε and a recently characterized CK1δ2 splice variant more efficiently prime mPER2 for downstream phosphorylation in cells than the well-studied splice variant CK1δ1. While CK1 phosphorylation of PER2 was previously shown to be robust to changes in the cellular environment, our phosphoswitch mathematical model of circadian rhythms shows that the CK1 carboxyl-terminal tail can allow the period of the clock to be sensitive to cellular signaling. These studies implicate the extreme carboxyl terminus of CK1 as a key regulator of circadian timing.


Subject(s)
Casein Kinase 1 epsilon/metabolism , Casein Kinase Idelta/metabolism , Circadian Rhythm/physiology , Period Circadian Proteins/metabolism , Animals , HEK293 Cells , Humans , Mice , Period Circadian Proteins/genetics , Phosphorylation
17.
Mol Pharmacol ; 97(2): 72-89, 2020 02.
Article in English | MEDLINE | ID: mdl-31787618

ABSTRACT

Wnts are secreted proteins that bind to cell surface receptors to activate downstream signaling cascades. Normal Wnt signaling plays key roles in embryonic development and adult tissue homeostasis. The secretion of Wnt ligands, the turnover of Wnt receptors, and the signaling transduction are tightly regulated and fine-tuned to keep the signaling output "just right." Hyperactivated Wnt signaling due to recurrent genetic alterations drives several human cancers. Elevated Wnt signaling also confers resistance to multiple conventional and targeted cancer therapies through diverse mechanisms including maintaining the cancer stem cell population, enhancing DNA damage repair, facilitating transcriptional plasticity, and promoting immune evasion. Different classes of Wnt signaling inhibitors targeting key nodes of the pathway have been developed and show efficacy in treating Wnt-driven cancers and subverting Wnt-mediated therapy resistance in preclinical studies. Several of these inhibitors have advanced to clinical trials, both singly and in combination with other existing US Food and Drug Administration-approved anti-cancer modalities. In the near future, pharmacological inhibition of Wnt signaling may be a real choice for patients with cancer. SIGNIFICANCE STATEMENT: The latest insights in Wnt signaling, ranging from basic biology to therapeutic implications in cancer, are reviewed. Recent studies extend understanding of this ancient signaling pathway and describe the development and improvement of anti-Wnt therapeutic modalities for cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis/genetics , Clinical Trials as Topic , DNA Damage/drug effects , DNA Repair/genetics , Drug Evaluation, Preclinical , Humans , Ligands , Molecular Targeted Therapy/methods , Mutation , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt Signaling Pathway/drug effects
18.
Differentiation ; 108: 8-16, 2019.
Article in English | MEDLINE | ID: mdl-30683451

ABSTRACT

Intestinal homeostasis is dependent on the continuous production of differentiated epithelial cells from a sustainable and resilient stem cell compartment. Wnt/ß-catenin signaling plays a central role in this process, cooperating with R-spondins, growth factors and regulators of the TGF-ß/BMP pathway to generate a specialized tissue microenvironment that regulates the intestinal stem cell niche. Recent studies revealed that many of these factors are produced in a paracrine manner by specialized cell populations that reside in the subepithelial stroma. These stromal signal-producing cells, variously called telocytes and myofibroblasts, can be identified by expression of specific genes including PdgfRa, Gli1 and FoxL1. In this review we discuss how the intestinal stem cell niche is established during development and then sustained during adult intestinal homeostasis by these stromal cell populations. The signaling stroma cells regulate intestinal stem cell development into different epithelial lineages and play an important role in the response to environmental stresses.


Subject(s)
Cell Self Renewal , Intestinal Mucosa/metabolism , Stem Cell Niche , Animals , Humans , Intestinal Mucosa/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Wnt Signaling Pathway
19.
Kidney Int ; 96(6): 1308-1319, 2019 12.
Article in English | MEDLINE | ID: mdl-31585741

ABSTRACT

Wnt/ß-catenin signaling is essential in the pathogenesis of renal fibrosis. We previously reported inhibition of the Wnt O-acyl transferase porcupine, required for Wnt secretion, dramatically attenuates kidney fibrosis in the murine unilateral ureteral obstruction model. Here, we investigated the tissue-specific contributions of porcupine to renal fibrosis and inflammation in ureteral obstruction using mice with porcupine deletion restricted to the kidney tubular epithelium or infiltrating myeloid cells. Obstruction of the ureter induced the renal mRNA expression of porcupine and downstream targets, ß-catenin, T-cell factor, and lymphoid enhancer factor in wild type mice. Renal tubular specific deficiency of porcupine reduced the expression of collagen I and other fibrosis markers in the obstructed kidney. Moreover, kidneys from obstructed mice with tubule-specific porcupine deficiency had reduced macrophage accumulation with attenuated expression of myeloid cytokine and chemokine mRNA. In co-culture with activated macrophages, renal tubular cells from tubular-specific porcupine knockout mice had blunted induction of fibrosis mediators compared with wild type renal tubular cells. In contrast, macrophages from macrophage-specific porcupine deficient mice in co-culture with wild type renal tubular cells had markedly enhanced expression of pro-fibrotic cytokines compared to wild type macrophages. Consequently, porcupine deletion specifically within macrophages augmented renal scar formation following ureteral obstruction. Thus, our experiments suggest a benefit of interrupting Wnt secretion specifically within the kidney epithelium while preserving Wnt O-acylation in infiltrating myeloid cells during renal fibrogenesis.


Subject(s)
Acyltransferases/metabolism , Membrane Proteins/metabolism , Nephrosclerosis/metabolism , Wnt Signaling Pathway , Animals , Chemokines/metabolism , Female , Fibrosis , Kidney Tubules/metabolism , Kidney Tubules/pathology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Myeloid Cells/metabolism , Nephrosclerosis/etiology , Ureteral Obstruction
20.
J Cell Sci ; 130(9): 1532-1544, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28289266

ABSTRACT

Wnt ligands are involved in diverse signaling pathways that are active during development, maintenance of tissue homeostasis and in various disease states. While signaling regulated by individual Wnts has been extensively studied, Wnts are rarely expressed alone, and the consequences of Wnt gene co-expression are not well understood. Here, we studied the effect of co-expression of Wnts on the ß-catenin signaling pathway. While some Wnts are deemed 'non-canonical' due to their limited ability to activate ß-catenin when expressed alone, unexpectedly, we find that multiple Wnt combinations can synergistically activate ß-catenin signaling in multiple cell types. WNT1- and WNT7B-mediated synergistic Wnt signaling requires FZD5, FZD8 and LRP6, as well as the WNT7B co-receptors GPR124 (also known as ADGRA2) and RECK. Unexpectedly, this synergistic signaling occurs downstream of ß-catenin stabilization, and is correlated with increased lysine acetylation of ß-catenin. Wnt synergy provides a general mechanism to confer increased combinatorial control over this important regulatory pathway.


Subject(s)
Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Acetylation , Clone Cells , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Models, Biological , Phosphorylation , Protein Stability , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , Stomach Neoplasms/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL