1
|
Martinez-Marin D, Stroman GC, Fulton CJ, Pruitt K. Frizzled receptors: gatekeepers of Wnt signaling in development and disease. Front Cell Dev Biol 2025; 13:1599355. [PMID: 40376615 PMCID: PMC12078226 DOI: 10.3389/fcell.2025.1599355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/21/2025] [Indexed: 05/18/2025] Open
Abstract
Frizzled (FZD) receptors are a subset of G-protein-coupled receptors (GPCRs), the largest class of human cell surface receptors and a major target of FDA-approved drugs. Activated by Wnt ligands, FZDs regulate key cellular processes such as proliferation, differentiation, and polarity, positioning them at the intersection of developmental biology and disease, including cancer. Despite their significance, FZD signaling remains incompletely understood, particularly in distinguishing receptor-specific roles across canonical and non-canonical Wnt pathways. Challenges include defining ligand-receptor specificity, elucidating signal transduction mechanisms, and understanding the influence of post translational modifications and the cellular context. Structural dynamics, receptor trafficking, and non-canonical signaling contributions also remain areas of active investigation. Recent advances in structural biology, transcriptomics, and functional genomics are beginning to address these gaps, while emerging therapeutic approaches-such as small-molecule modulators and antibodies-highlight the potential of FZDs as drug targets. This review synthesizes current insights into FZD receptor biology, examines ongoing controversies, and outlines promising directions for future research and therapeutic development.
Collapse
Affiliation(s)
| | | | | | - Kevin Pruitt
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
2
|
Schubert A, Mongkolsittisilp A, Kobitski A, Schulz M, Voloshanenko O, Schaffrinski M, Winkler N, Neßling M, Richter K, Kranz D, Nienhaus K, Jäger D, Trümper L, Büntzel J, Binder C, Nienhaus GU, Boutros M. WNT5a export onto extracellular vesicles studied at single-molecule and single-vesicle resolution. FEBS J 2025. [PMID: 40165582 DOI: 10.1111/febs.70074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 11/17/2024] [Accepted: 01/10/2025] [Indexed: 04/02/2025]
Abstract
WNT signaling governs development, homeostasis, and aging of cells and tissues, and is frequently dysregulated in pathophysiological processes such as cancer. WNT proteins are hydrophobic and traverse the intercellular space between the secreting and receiving cells on various carriers, including extracellular vesicles (EVs). Here, we address the relevance of different EV fractions and other vehicles for WNT5a protein, a non-canonical WNT ligand that signals independently of beta-catenin. Its highly context-dependent roles in cancer (either tumor-suppressive or tumor-promoting) have been attributed to two distinct isoforms, WNT5a Short (WNT5aS) and WNT5a Long (WNT5aL), resulting from different signal peptide cleavage sites. To explore possible differences in secretion and extracellular transport, we developed fusion constructs with the fluorescent proteins (FPs) mScarlet and mOxNeonGreen. Functional reporter assays revealed that both WNT5a isoforms inhibit canonical WNT signaling, and EVs produced by WNT5a-bearing tumor cells, carrying either of the WNT5a isoforms, induced invasiveness of the luminal A breast cancer cell line MCF7. We used fluorescence intensity distribution analysis (FIDA) and fluorescence correlation spectroscopy (FCS) to characterize at single-molecule sensitivity WNT5aL-bearing entities secreted by HEK293T cells. Importantly, we found that most WNT5aL proteins remained monomeric in the supernatant after ultracentrifugation; only a minor fraction was EV-bound. We further determined the average sizes of the EV fractions and the average number of WNT5aL proteins per EV. Our detailed biophysical analysis of the physical nature of the EV populations is an important step toward understanding context-dependent WNT cargo loading and signaling in future studies.
Collapse
Affiliation(s)
- Antonia Schubert
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute for Human Genetics, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Germany
| | | | - Andrei Kobitski
- Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Matthias Schulz
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Germany
| | - Oksana Voloshanenko
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute for Human Genetics, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Meike Schaffrinski
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Germany
| | - Nadine Winkler
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute for Human Genetics, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Michelle Neßling
- Central Unit Electron Microscopy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karsten Richter
- Central Unit Electron Microscopy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominique Kranz
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Germany
| | - Lorenz Trümper
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Germany
| | - Judith Büntzel
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Germany
| | - Claudia Binder
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Germany
| | - Gerd Ulrich Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany
- Institute of Nanotechnology, Karlsruhe Institute of Technology, Karlsruhe, Germany
- Institute of Biological and Chemical Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Michael Boutros
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Institute for Human Genetics, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
3
|
Bruguera ES, Mahoney JP, Weis WI. The co-receptor Tetraspanin12 directly captures Norrin to promote ligand-specific β-catenin signaling. eLife 2025; 13:RP96743. [PMID: 39745873 DOI: 10.7554/elife.96743] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Wnt/β-catenin signaling directs animal development and tissue renewal in a tightly controlled, cell- and tissue-specific manner. In the mammalian central nervous system, the atypical ligand Norrin controls angiogenesis and maintenance of the blood-brain barrier and blood-retina barrier through the Wnt/β-catenin pathway. Like Wnt, Norrin activates signaling by binding and heterodimerizing the receptors Frizzled (Fzd) and low-density lipoprotein receptor-related protein 5 or 6 (LRP5/6), leading to membrane recruitment of the intracellular transducer Dishevelled (Dvl) and ultimately stabilizing the transcriptional coactivator β-catenin. Unlike Wnt, the cystine knot ligand Norrin only signals through Fzd4 and additionally requires the co-receptor Tetraspanin12 (Tspan12); however, the mechanism underlying Tspan12-mediated signal enhancement is unclear. It has been proposed that Tspan12 integrates into the Norrin-Fzd4 complex to enhance Norrin-Fzd4 affinity or otherwise allosterically modulate Fzd4 signaling. Here, we measure direct, high-affinity binding between purified Norrin and Tspan12 in a lipid environment and use AlphaFold models to interrogate this interaction interface. We find that Tspan12 and Fzd4 can simultaneously bind Norrin and that a pre-formed Tspan12/Fzd4 heterodimer, as well as cells co-expressing Tspan12 and Fzd4, more efficiently capture low concentrations of Norrin than Fzd4 alone. We also show that Tspan12 competes with both heparan sulfate proteoglycans and LRP6 for Norrin binding and that Tspan12 does not impact Fzd4-Dvl affinity in the presence or absence of Norrin. Our findings suggest that Tspan12 does not allosterically enhance Fzd4 binding to Norrin or Dvl, but instead functions to directly capture Norrin upstream of signaling.
Collapse
Affiliation(s)
- Elise S Bruguera
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, United States
| | - Jacob P Mahoney
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, United States
| | - William I Weis
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
4
|
Zheng G, Lin S, Wang S, Yan Y, Zheng D. Regulation of Natural Products on Wnt/β-Catenin Signaling Pathway in Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:709-735. [PMID: 40374374 DOI: 10.1142/s0192415x25500272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
The Wnt/β-catenin signaling pathway plays a crucial role in both physiological and pathological conditions. Targeting molecules associated with the Wnt/β-catenin signaling pathway presents a promising approach for disease treatment. The use of natural products in treating various diseases is widespread due to their favorable biocompatibility, low toxicity, and high biological activity. Research has shown that natural products such as curcumin and resveratrol can regulate multiple signaling pathways under disease conditions, including the Wnt/β-catenin signaling pathway. However, the regulatory mechanisms of natural products remain incompletely understood. This review aims to explore the regulatory effects of natural products on the Wnt/β-catenin signaling pathway in certain diseases, especially in the process of tumor progression. It outlines the composition and mechanisms of the Wnt/β-catenin signaling pathway. Furthermore, we predicted the potential binding sites of these natural products to this pathway, summarized the effects of diverse natural products on this signaling pathway, and conducted a preliminary exploration ofd the mechanisms of the effects of natural products. In addition, we considered and discussed the limitations of natural products, such as potential side effects from long-term use and the precision in targeting the Wnt/β-catenin signaling pathway. This review provides a theoretical basis for the targeted strategy of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Genggeng Zheng
- School of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shuoqi Lin
- School of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shijie Wang
- School of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yuxiang Yan
- School of Stomatology, Fujian Medical University, Fuzhou, China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
5
|
Koval A, Boudou C, Katanaev VL. Challenging Reported Frizzled-Targeting Compounds in Selective Assays Reveals Lack of Functional Inhibition and Claimed Profiles. ACS Pharmacol Transl Sci 2024; 7:4144-4154. [PMID: 39698282 PMCID: PMC11650735 DOI: 10.1021/acsptsci.4c00570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Selective inhibitors of Frizzled (FZD) GPCRs are highly sought after as potentially highly efficacious and safe treatments for cancer as well as tools in regenerative medicine and fundamental science. In recent years, there have been several reports claiming the identification of small molecule agents that are selective toward certain FZD proteins using a variety of approaches. However, the majority of these studies lacked a selective functional assay to validate their functionality. In this study, we describe the development and application of a selective assay for individual FZD proteins. Our findings indicate that the majority of reported compounds lack the capacity to inhibit the functioning of the claimed FZD proteins when stimulated by a Wnt ligand in the canonical pathway. Instead, the compounds demonstrate a broad range of off-target effects, including inhibition of downstream pathway component(s) (3235-0367, SRI35959, carbamazepine, niclosamide), lack of activity (FzM1), and surprising antagonism of firefly luciferase (F7H). The only compound that fulfills the expected selectivity profile is peptide Fz7-21. These results highlight the necessity of implementing rigorous testing of the screening-derived compounds in selective functional assays and are important for the field of drug discovery and development targeting the highly demanded Wnt-FZD pathway.
Collapse
Affiliation(s)
- Alexey Koval
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| | - Cédric Boudou
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| | - Vladimir L. Katanaev
- Department of Cell Physiology
and Metabolism, Translational Research Centre in Oncohaematology,
Faculty of Medicine, University of Geneva,1206 Geneva, Switzerland
| |
Collapse
|
6
|
Bruguera ES, Mahoney JP, Weis WI. The co-receptor Tetraspanin12 directly captures Norrin to promote ligand-specific β-catenin signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.03.578714. [PMID: 38352533 PMCID: PMC10862866 DOI: 10.1101/2024.02.03.578714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Wnt/β-catenin signaling directs animal development and tissue renewal in a tightly controlled, cell- and tissue-specific manner. In the mammalian central nervous system, the atypical ligand Norrin controls angiogenesis and maintenance of the blood-brain barrier and blood-retina barrier through the Wnt/β-catenin pathway. Like Wnt, Norrin activates signaling by binding and heterodimerizing the receptors Frizzled (Fzd) and Low-density lipoprotein receptor-related protein 5 or 6 (LRP5/6), leading to membrane recruitment of the intracellular transducer Dishevelled (Dvl) and ultimately stabilizing the transcriptional coactivator β-catenin. Unlike Wnt, the cystine-knot ligand Norrin only signals through Fzd4 and additionally requires the co-receptor Tetraspanin12 (Tspan12); however, the mechanism underlying Tspan12-mediated signal enhancement is unclear. It has been proposed that Tspan12 integrates into the Norrin-Fzd4 complex to enhance Norrin-Fzd4 affinity or otherwise allosterically modulate Fzd4 signaling. Here, we measure direct, high-affinity binding between purified Norrin and Tspan12 in a lipid environment and use AlphaFold models to interrogate this interaction interface. We find that Tspan12 and Fzd4 can simultaneously bind Norrin and that a pre-formed Tspan12/Fzd4 heterodimer, as well as cells co-expressing Tspan12 and Fzd4, more efficiently capture low concentrations of Norrin than Fzd4 alone. We also show that Tspan12 competes with both heparan sulfate proteoglycans and LRP6 for Norrin binding and that Tspan12 does not impact Fzd4-Dvl affinity in the presence or absence of Norrin. Our findings suggest that Tspan12 does not allosterically enhance Fzd4 binding to Norrin or Dvl, but instead functions to directly capture Norrin upstream of signaling.
Collapse
Affiliation(s)
- Elise S Bruguera
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Jacob P Mahoney
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - William I Weis
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
7
|
Ensing J, Ide AD, Gilliland C, Tsurho V, Caza I, Stratman AN, Lanning NJ, Grainger S. The E3 Ubiquitin Ligase Trip12 attenuates Wnt9a/Fzd9b signaling during hematopoietic stem cell development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620301. [PMID: 39484584 PMCID: PMC11527353 DOI: 10.1101/2024.10.25.620301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Wnt signaling is essential for both the development and homeostasis of diverse cellular lineages, including hematopoietic stem cells. Organism-wide, Wnt signals are tightly regulated, as overactivation of the pathway can lead to tumorigenesis. Although numerous Wnt ligands and Frizzled (Fzd) receptors exist, how particular Wnt/Fzd pairings are established and how their signals are regulated is poorly understood. We have previously identified the requirements of the cognate pairing of Wnt9a and Fzd9b for early hematopoietic stem cell proliferation. However, the specific signals governing activation, but equally important, the molecular mechanisms required to turn the signal 'off,' are unknown. Here, we show that the E3 ubiquitin ligase Trip12 (thyroid hormone receptor interactor 12) is specifically required to ubiquitinate the third intracellular loop of Fzd9b at K437, targeting it for lysosomal degradation. In contrast to other ubiquitin ligases described to regulate the cell surface availability of multiple Fzds broadly, our data indicate that Trip12 is selective for Fzd9b. We further demonstrate that this occurs through ubiquitination at K437 of Fzd9b in the third intracellular loop, ultimately leading to a decrease in Fzd9b receptor availability and in Wnt9a/Fzd9b signaling that impacts hematopoietic stem cell proliferation in zebrafish. Our results point to specific mechanisms driving the availability of different Fzd receptors. Determining how particular Fzd abundance is regulated at the membrane will be critical to developing specific therapies for human intervention.
Collapse
Affiliation(s)
- Jessica Ensing
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Amber D. Ide
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Carla Gilliland
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Visakuo Tsurho
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Isabella Caza
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Amber N. Stratman
- Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA RRID: SCR_000343
| | - Nathan J. Lanning
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| |
Collapse
|
8
|
Schulte G. International Union of Basic and Clinical Pharmacology CXV: The Class F of G Protein-Coupled Receptors. Pharmacol Rev 2024; 76:1009-1037. [PMID: 38955509 DOI: 10.1124/pharmrev.124.001062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/10/2024] [Accepted: 05/17/2024] [Indexed: 07/04/2024] Open
Abstract
The class F of G protein-coupled receptors (GPCRs) consists of 10 Frizzleds (FZD1-10) and Smoothened (SMO). FZDs bind and are activated by secreted lipoglycoproteins of the Wingless/Int-1 (WNT) family, and SMO is indirectly activated by the Hedgehog (Hh) family of morphogens acting on the transmembrane protein Patched. The advance of our understanding of FZDs and SMO as dynamic transmembrane receptors and molecular machines, which emerged during the past 14 years since the first-class F GPCR IUPHAR nomenclature report, justifies an update. This article focuses on the advances in molecular pharmacology and structural biology providing new mechanistic insight into ligand recognition, receptor activation mechanisms, signal initiation, and signal specification. Furthermore, class F GPCRs continue to develop as drug targets, and novel technologies and tools such as genetically encoded biosensors and CRISP/Cas9 edited cell systems have contributed to refined functional analysis of these receptors. Also, advances in crystal structure analysis and cryogenic electron microscopy contribute to the rapid development of our knowledge about structure-function relationships, providing a great starting point for drug development. Despite the progress, questions and challenges remain to fully understand the complexity of the WNT/FZD and Hh/SMO signaling systems. SIGNIFICANCE STATEMENT: The recent years of research have brought about substantial functional and structural insight into mechanisms of activation of Frizzleds and Smoothened. While the advance furthers our mechanistic understanding of ligand recognition, receptor activation, signal specification, and initiation, broader opportunities emerge that allow targeting class F GPCRs for therapy and regenerative medicine employing both biologics and small molecule compounds.
Collapse
Affiliation(s)
- Gunnar Schulte
- Karolinska Institutet, Department of Physiology & Pharmacology, Receptor Biology & Signaling, Biomedicum, Stockholm, Sweden
| |
Collapse
|
9
|
Grätz L, Voss JH, Schulte G. Class-Wide Analysis of Frizzled-Dishevelled Interactions Using BRET Biosensors Reveals Functional Differences among Receptor Paralogs. ACS Sens 2024; 9:4626-4636. [PMID: 39213612 PMCID: PMC11443525 DOI: 10.1021/acssensors.4c00806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Wingless/Int-1 (WNT) signaling is mediated by WNT binding to 10 Frizzleds (FZD1-10), which propagate the signal inside the cell by interacting with different transducers, most prominently the phosphoprotein Dishevelled (DVL). Despite recent progress, questions about WNT/FZD selectivity and paralog-dependent differences in the FZD/DVL interaction remain unanswered. Here, we present a class-wide analysis of the FZD/DVL interaction using the DEP domain of DVL as a proxy in bioluminescence resonance energy transfer (BRET) techniques. Most FZDs engage in a constitutive high-affinity interaction with DEP. Stimulation of unimolecular FZD/DEP BRET sensors with different ligands revealed that most paralogs are dynamic in the FZD/DEP interface, showing distinct profiles in terms of ligand selectivity and signal kinetics. This study underlines mechanistic differences in terms of how allosteric communication between FZDs and their main signal transducer DVL occurs. Moreover, the unimolecular sensors represent the first receptor-focused biosensors to surpass the requirements for high-throughput screening, facilitating FZD-targeted drug discovery.
Collapse
Affiliation(s)
- Lukas Grätz
- Department of Physiology & Pharmacology, Section of Receptor Biology & Signaling, Biomedicum, Karolinska Institutet, S-17165 Stockholm, Sweden
| | - Jan H Voss
- Department of Physiology & Pharmacology, Section of Receptor Biology & Signaling, Biomedicum, Karolinska Institutet, S-17165 Stockholm, Sweden
| | - Gunnar Schulte
- Department of Physiology & Pharmacology, Section of Receptor Biology & Signaling, Biomedicum, Karolinska Institutet, S-17165 Stockholm, Sweden
| |
Collapse
|
10
|
Xie N, Landin Malt A, Adylkhan A, Rodeman N, Moraes Borges R, Hwang D, Liu A, Smith C, Hogan A, Lu X. Wnt7b acts in concert with Wnt5a to regulate tissue elongation and planar cell polarity via noncanonical Wnt signaling. Proc Natl Acad Sci U S A 2024; 121:e2405217121. [PMID: 39172791 PMCID: PMC11363310 DOI: 10.1073/pnas.2405217121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024] Open
Abstract
Intercellular signaling mediated by evolutionarily conserved planar cell polarity (PCP) proteins aligns cell polarity along the tissue plane and drives polarized cell behaviors during tissue morphogenesis. Accumulating evidence indicates that the vertebrate PCP pathway is regulated by noncanonical, β-catenin-independent Wnt signaling; however, the signaling components and mechanisms are incompletely understood. In the mouse hearing organ, both PCP and noncanonical Wnt (ncWnt) signaling are required in the developing auditory sensory epithelium to control cochlear duct elongation and planar polarity of resident sensory hair cells (HCs), including the shape and orientation of the stereociliary hair bundle essential for sound detection. We have recently discovered a Wnt/G-protein/PI3K pathway that coordinates HC planar polarity and intercellular PCP signaling. Here, we identify Wnt7b as a ncWnt ligand acting in concert with Wnt5a to promote tissue elongation in diverse developmental processes. In the cochlea, Wnt5a and Wnt7b are redundantly required for cochlear duct coiling and elongation, HC planar polarity, and asymmetric localization of core PCP proteins Fzd6 and Dvl2. Mechanistically, Wnt5a/Wnt7b-mediated ncWnt signaling promotes membrane recruitment of Daple, a nonreceptor guanine nucleotide exchange factor for Gαi, and activates PI3K/AKT and ERK signaling, which promote asymmetric Fzd6 localization. Thus, ncWnt and PCP signaling pathways have distinct mutant phenotypes and signaling components, suggesting that they act as separate, parallel pathways with nonoverlapping functions in cochlear morphogenesis. NcWnt signaling drives tissue elongation and reinforces intercellular PCP signaling by regulating the trafficking of PCP-specific Frizzled receptors.
Collapse
Affiliation(s)
- Nicholas Xie
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Andre Landin Malt
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Aray Adylkhan
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Natalie Rodeman
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Ricardo Moraes Borges
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Diane Hwang
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Alice Liu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Connor Smith
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Arielle Hogan
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| | - Xiaowei Lu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA22908
| |
Collapse
|
11
|
Ahmad S, Attisano L. Wnt5a Promotes Axon Elongation in Coordination with the Wnt-Planar Cell Polarity Pathway. Cells 2024; 13:1268. [PMID: 39120298 PMCID: PMC11312420 DOI: 10.3390/cells13151268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
The establishment of neuronal polarity, involving axon specification and outgrowth, is critical to achieve the proper morphology of neurons, which is important for neuronal connectivity and cognitive functions. Extracellular factors, such as Wnts, modulate diverse aspects of neuronal morphology. In particular, non-canonical Wnt5a exhibits differential effects on neurite outgrowth depending upon the context. Thus, the role of Wnt5a in axon outgrowth and neuronal polarization is not completely understood. In this study, we demonstrate that Wnt5a, but not Wnt3a, promotes axon outgrowth in dissociated mouse embryonic cortical neurons and does so in coordination with the core PCP components, Prickle and Vangl. Unexpectedly, exogenous Wnt5a-induced axon outgrowth was dependent on endogenous, neuronal Wnts, as the chemical inhibition of Porcupine using the IWP2- and siRNA-mediated knockdown of either Porcupine or Wntless inhibited Wnt5a-induced elongation. Importantly, delayed treatment with IWP2 did not block Wnt5a-induced elongation, suggesting that endogenous Wnts and Wnt5a act during specific timeframes of neuronal polarization. Wnt5a in fibroblast-conditioned media can associate with small extracellular vesicles (sEVs), and we also show that these Wnt5a-containing sEVs are primarily responsible for inducing axon elongation.
Collapse
Affiliation(s)
| | - Liliana Attisano
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada;
| |
Collapse
|
12
|
Tümen D, Heumann P, Huber J, Hahn N, Macek C, Ernst M, Kandulski A, Kunst C, Gülow K. Unraveling Cancer's Wnt Signaling: Dynamic Control through Protein Kinase Regulation. Cancers (Basel) 2024; 16:2686. [PMID: 39123414 PMCID: PMC11312265 DOI: 10.3390/cancers16152686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Since the initial identification of oncogenic Wnt in mice and Drosophila, the Wnt signaling pathway has been subjected to thorough and extensive investigation. Persistent activation of Wnt signaling exerts diverse cancer characteristics, encompassing tumor initiation, tumor growth, cell senescence, cell death, differentiation, and metastasis. Here we review the principal signaling mechanisms and the regulatory influence of pathway-intrinsic and extrinsic kinases on cancer progression. Additionally, we underscore the divergences and intricate interplays of the canonical and non-canonical Wnt signaling pathways and their critical influence in cancer pathophysiology, exhibiting both growth-promoting and growth-suppressing roles across diverse cancer types.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Karsten Gülow
- Department of Internal Medicine I Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (D.T.); (N.H.)
| |
Collapse
|
13
|
Nguyen N, Carpenter KA, Ensing J, Gilliland C, Rudisel EJ, Mu EM, Thurlow KE, Triche TJ, Grainger S. EGFR-dependent endocytosis of Wnt9a and Fzd9b promotes β-catenin signaling during hematopoietic stem cell development in zebrafish. Sci Signal 2024; 17:eadf4299. [PMID: 38626007 PMCID: PMC11103623 DOI: 10.1126/scisignal.adf4299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/28/2024] [Indexed: 04/18/2024]
Abstract
Cell-to-cell communication through secreted Wnt ligands that bind to members of the Frizzled (Fzd) family of transmembrane receptors is critical for development and homeostasis. Wnt9a signals through Fzd9b, the co-receptor LRP5 or LRP6 (LRP5/6), and the epidermal growth factor receptor (EGFR) to promote early proliferation of zebrafish and human hematopoietic stem cells during development. Here, we developed fluorescently labeled, biologically active Wnt9a and Fzd9b fusion proteins to demonstrate that EGFR-dependent endocytosis of the ligand-receptor complex was required for signaling. In human cells, the Wnt9a-Fzd9b complex was rapidly endocytosed and trafficked through early and late endosomes, lysosomes, and the endoplasmic reticulum. Using small-molecule inhibitors and genetic and knockdown approaches, we found that Wnt9a-Fzd9b endocytosis required EGFR-mediated phosphorylation of the Fzd9b tail, caveolin, and the scaffolding protein EGFR protein substrate 15 (EPS15). LRP5/6 and the downstream signaling component AXIN were required for Wnt9a-Fzd9b signaling but not for endocytosis. Knockdown or loss of EPS15 impaired hematopoietic stem cell development in zebrafish. Other Wnt ligands do not require endocytosis for signaling activity, implying that specific modes of endocytosis and trafficking may represent a method by which Wnt-Fzd specificity is established.
Collapse
Affiliation(s)
- Nicole Nguyen
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID:SCR_021956
| | - Kelsey A. Carpenter
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID:SCR_021956
| | - Jessica Ensing
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID:SCR_021956
| | - Carla Gilliland
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID:SCR_021956
| | - Emma J. Rudisel
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID:SCR_021956
| | - Emily M. Mu
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID:SCR_021956
| | - Kate E. Thurlow
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID:SCR_021956
- Van Andel Institute Graduate School, Grand Rapids, Michigan, 49503, USA
| | - Timothy J. Triche
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan, 49503, USA
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID:SCR_021956
| |
Collapse
|
14
|
Massenet-Regad L, Soumelis V. ICELLNET v2: a versatile method for cell-cell communication analysis from human transcriptomic data. Bioinformatics 2024; 40:btae089. [PMID: 38490248 PMCID: PMC10955248 DOI: 10.1093/bioinformatics/btae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/31/2024] [Accepted: 03/13/2024] [Indexed: 03/17/2024] Open
Abstract
SUMMARY Several methods have been developed in the past years to infer cell-cell communication networks from transcriptomic data based on ligand and receptor expression. Among them, ICELLNET is one of the few approaches to consider the multiple subunits of ligands and receptors complexes to infer and quantify cell communication. In here, we present a major update of ICELLNET. As compared to its original implementation, we (i) drastically expanded the ICELLNET ligand-receptor database from 380 to 1669 biologically curated interactions, (ii) integrated important families of communication molecules involved in immune crosstalk, cell adhesion, and Wnt pathway, (iii) optimized ICELLNET framework for single-cell RNA sequencing data analyses, (iv) provided new visualizations of cell-cell communication results to facilitate prioritization and biological interpretation. This update will broaden the use of ICELLNET by the scientific community in different biological fields. AVAILABILITY AND IMPLEMENTATION ICELLNET package is implemented in R. Source code, documentation and tutorials are available on GitHub (https://github.com/soumelis-lab/ICELLNET).
Collapse
Affiliation(s)
- Lucile Massenet-Regad
- Université Paris Cité, INSERM U976 HIPI, Paris, F-75010, France
- Université Paris-Saclay, Saint Aubin, F-91190, France
| | - Vassili Soumelis
- Université Paris Cité, INSERM U976 HIPI, Paris, F-75010, France
- Department of Immunology-Histocompatibility, Saint-Louis Hospital, AP-HP.Nord, Université Paris Cité, Paris 75010, France
- Owkin France, Paris 75010, France
| |
Collapse
|
15
|
Liu Y, Sun J, Zhang C, Wu Y, Ma S, Li X, Wu X, Gao Q. Compound heterozygous WNT10A missense variations exacerbated the tooth agenesis caused by hypohidrotic ectodermal dysplasia. BMC Oral Health 2024; 24:136. [PMID: 38280992 PMCID: PMC10822191 DOI: 10.1186/s12903-024-03888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 01/12/2024] [Indexed: 01/29/2024] Open
Abstract
BACKGROUND The aim of this study was to analyse the differences in the phenotypes of missing teeth between a pair of brothers with hypohidrotic ectodermal dysplasia (HED) and to investigate the underlying mechanism by comparing the mutated gene loci between the brothers with whole-exome sequencing. METHODS The clinical data of the patients and their mother were collected, and genomic DNA was extracted from peripheral blood samples. By Whole-exome sequencing filtered for a minor allele frequency (MAF) ≤0.05 non-synonymous single-nucleotide variations and insertions/deletions variations in genes previously associated with tooth agenesis, and variations considered as potentially pathogenic were assessed by SIFT, Polyphen-2, CADD and ACMG. Sanger sequencing was performed to detect gene variations. The secondary and tertiary structures of the mutated proteins were predicted by PsiPred 4.0 and AlphaFold 2. RESULTS Both brothers were clinically diagnosed with HED, but the younger brother had more teeth than the elder brother. An EDA variation (c.878 T > G) was identified in both brothers. Additionally, compound heterozygous variations of WNT10A (c.511C > T and c.637G > A) were identified in the elder brother. Digenic variations in EDA (c.878 T > G) and WNT10A (c.511C > T and c.637G > A) in the same patient have not been reported previously. The secondary structure of the variant WNT10A protein showed changes in the number and position of α-helices and β-folds compared to the wild-type protein. The tertiary structure of the WNT10A variant and molecular simulation docking showed that the site and direction where WNT10A binds to FZD5 was changed. CONCLUSIONS Compound heterozygous WNT10A missense variations may exacerbate the number of missing teeth in HED caused by EDA variation.
Collapse
Affiliation(s)
- Yiting Liu
- The Stomatology Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Academician Workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha, Hunan Province, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jing Sun
- The Stomatology Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Academician Workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha, Hunan Province, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Caiqi Zhang
- The Stomatology Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Academician Workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha, Hunan Province, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yi Wu
- The Stomatology Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Academician Workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha, Hunan Province, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Siyuan Ma
- The Stomatology Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Academician Workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha, Hunan Province, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xuechun Li
- The Stomatology Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Academician Workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha, Hunan Province, China
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiaoshan Wu
- The Stomatology Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- Academician Workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha, Hunan Province, China.
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.
| | - Qingping Gao
- The Stomatology Center of Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- Academician Workstation for Oral & Maxillofacial Regenerative Medicine, Central South University, Changsha, Hunan Province, China.
- Research Center of Oral and Maxillofacial Development and Regeneration, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
16
|
Granados AA, Kanrar N, Elowitz MB. Combinatorial expression motifs in signaling pathways. CELL GENOMICS 2024; 4:100463. [PMID: 38216284 PMCID: PMC10794782 DOI: 10.1016/j.xgen.2023.100463] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 08/02/2023] [Accepted: 11/15/2023] [Indexed: 01/14/2024]
Abstract
In animal cells, molecular pathways often comprise families of variant components, such as ligands or receptors. These pathway components are differentially expressed by different cell types, potentially tailoring pathway function to cell context. However, it has remained unclear how pathway expression profiles are distributed across cell types and whether similar profiles can occur in dissimilar cell types. Here, using single-cell gene expression datasets, we identified pathway expression motifs, defined as recurrent expression profiles that are broadly distributed across diverse cell types. Motifs appeared in core pathways, including TGF-β, Notch, Wnt, and the SRSF splice factors, and involved combinatorial co-expression of multiple components. Motif usage was weakly correlated between pathways in adult cell types and during dynamic developmental transitions. Together, these results suggest a mosaic view of cell type organization, in which different cell types operate many of the same pathways in distinct modes.
Collapse
Affiliation(s)
- Alejandro A Granados
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute and Department of Applied Physics, California Institute of Technology, Pasadena, CA 91125, USA
| | - Nivedita Kanrar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute and Department of Applied Physics, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute and Department of Applied Physics, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
17
|
Kulkarni PP, Ekhlak M, Dash D. Non-canonical non-genomic morphogen signaling in anucleate platelets: a critical determinant of prothrombotic function in circulation. Cell Commun Signal 2024; 22:13. [PMID: 38172855 PMCID: PMC10763172 DOI: 10.1186/s12964-023-01448-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Circulating platelets derived from bone marrow megakaryocytes play a central role in thrombosis and hemostasis. Despite being anucleate, platelets express several proteins known to have nuclear niche. These include transcription factors and steroid receptors whose non-genomic functions are being elucidated in platelets. Quite remarkably, components of some of the best-studied morphogen pathways, namely Notch, Sonic Hedgehog (Shh), and Wnt have also been described in recent years in platelets, which regulate platelet function in the context of thrombosis as well as influence their survival. Shh and Notch pathways in stimulated platelets establish feed-forward loops of autocrine/juxtacrine/paracrine non-canonical signaling that helps perpetuate thrombosis. On the other hand, non-canonical Wnt signaling is part of a negative feedback loop for restricting platelet activation and possibly limiting thrombus growth. The present review will provide an overview of these signaling pathways in general. We will then briefly discuss the non-genomic roles of transcription factors and steroid receptors in platelet activation. This will be followed by an elaborate description of morphogen signaling in platelets with a focus on their bearing on platelet activation leading to hemostasis and thrombosis as well as their potential for therapeutic targeting in thrombotic disorders.
Collapse
Affiliation(s)
- Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
18
|
Li N, Ge Q, Guo Q, Tao Y. Identification and functional validation of FZD8-specific antibodies. Int J Biol Macromol 2024; 254:127846. [PMID: 37926311 DOI: 10.1016/j.ijbiomac.2023.127846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
The Wnt pathway is an evolutionarily conserved pathway involved in stem cell homeostasis and tissue regeneration. Aberrant signaling in the Wnt pathway is highly associated with cancer. Developing antibodies to block overactivation of Frizzled receptors (FZDs), the main receptors in the Wnt pathway, is one of the viable options for treating cancer. However, obtaining isoform-specific antibodies is often challenging due to the high degree of homology among the ten FZDs. In this study, by using a synthetic library, we identified an antibody named pF8_AC3 that preferentially binds to FZD8. Guided by the structure of the complex of pF8_AC3 and FZD8, a second-generation targeted library was further constructed, and finally, the FZD8-specific antibody sF8_AG6 was obtained. Cell-based assays showed that these antibodies could selectively block FZD8-mediated signaling activation. Taken together, these antibodies have the potential to be developed into therapeutic drugs in the future.
Collapse
Affiliation(s)
- Na Li
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Qiangqiang Ge
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Qiong Guo
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| | - Yuyong Tao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Research Center for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
19
|
Alsaadi A, Artibani M, Hu Z, Wietek N, Morotti M, Gonzalez LS, Alazzam M, Jiang J, Abdul B, Soleymani Majd H, Blazer LL, Adams J, Silvestri F, Sidhu SS, Brugge JS, Ahmed AA. Single-cell transcriptomics identifies a WNT7A-FZD5 signaling axis that maintains fallopian tube stem cells in patient-derived organoids. Cell Rep 2023; 42:113354. [PMID: 37917586 DOI: 10.1016/j.celrep.2023.113354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/31/2023] [Accepted: 10/11/2023] [Indexed: 11/04/2023] Open
Abstract
The study of fallopian tube (FT) function in health and disease has been hampered by limited knowledge of FT stem cells and lack of in vitro models of stem cell renewal and differentiation. Using optimized organoid culture conditions to address these limitations, we find that FT stem cell renewal is highly dependent on WNT/β-catenin signaling and engineer endogenous WNT/β-catenin signaling reporter organoids to biomark, isolate, and characterize these cells. Using functional approaches, as well as bulk and single-cell transcriptomics analyses, we show that an endogenous hormonally regulated WNT7A-FZD5 signaling axis is critical for stem cell renewal and that WNT/β-catenin pathway-activated cells form a distinct transcriptomic cluster of FT cells enriched in extracellular matrix (ECM) remodeling and integrin signaling pathways. Overall, we provide a deep characterization of FT stem cells and their molecular requirements for self-renewal, paving the way for mechanistic work investigating the role of stem cells in FT health and disease.
Collapse
Affiliation(s)
- Abdulkhaliq Alsaadi
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Mara Artibani
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Zhiyuan Hu
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Nina Wietek
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Matteo Morotti
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Laura Santana Gonzalez
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Moiad Alazzam
- Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Jason Jiang
- Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Beena Abdul
- Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK
| | - Hooman Soleymani Majd
- Medical Sciences Division, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Levi L Blazer
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Jarret Adams
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | | | - Sachdev S Sidhu
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Joan S Brugge
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford OX3 9DU, UK; Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford OX3 7LE, UK.
| |
Collapse
|
20
|
Zhang X, Chang M, Wang B, Liu X, Zhang Z, Han G. YAP/WNT5A/FZD4 axis regulates osteogenic differentiation of human periodontal ligament cells under cyclic stretch. J Periodontal Res 2023; 58:907-918. [PMID: 37340863 DOI: 10.1111/jre.13143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/07/2023] [Accepted: 05/15/2023] [Indexed: 06/22/2023]
Abstract
OBJECTIVE To verify the role of YAP/WNT5A/FZD4 axis in stretch-induced osteogenic differentiation of hPDLCs. BACKGROUND During orthodontic tooth movement, differentiation of human periodontal ligament cells (hPDLCs) at the tension side of the periodontal ligament mediates new bone formation. WNT5A promotes osteogenesis and its regulator Yes-associated protein (YAP) is responsive to mechanical stimulation in hPDLCs. However, the mechanisms of YAP and WNT5A in alveolar bone remodeling remain unclear. METHODS Cyclic stretch was applied to hPDLCs to mimic the orthodontic stretching force. Osteogenic differentiation was determined by alkaline phosphatase (ALP) activity, Alizarin Red staining, qRT-PCR and western blotting. To detect activation of YAP and expression of WNT5A and its receptor Frizzled-4 (FZD4), western blotting, immunofluorescence, qRT-PCR and ELISA were performed. Verteporfin, Lats-IN-1, small interfering RNAs and recombinant protein were used to explore the relationship of YAP, WNT5A and FZD4, and the effect of their relationship on stretch-induced osteogenesis of hPDLCs. RESULTS WNT5A, FZD4 and nuclear localization of YAP were upregulated by cyclic stretch. YAP positively regulated WNT5A and FZD4 expression and osteogenic differentiation of hPDLCs under cyclic stretch by YAP inhibition or activation assay. Knockdown of WNT5A and FZD4 attenuated YAP-induced and stretch-induced osteogenic differentiation. Recombinant WNT5A rescued the suppressed osteogenic differentiation by YAP inhibitor in hPDLCs, whereas knockdown of FZD4 weakened the effect of WNT5A and amplified the suppression. CONCLUSIONS WNT5A/FZD4 could be positively regulated by YAP and the YAP/WNT5A/FZD4 axis mediated osteogenic differentiation of hPDLCs under cyclic stretch. This study provided further insight into the biological mechanism of orthodontic tooth movement.
Collapse
Affiliation(s)
- Xiaocen Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Maolin Chang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Orthodontic Department Division II, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Beike Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Orthodontic Department Division II, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiaoyu Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhen Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Orthodontic Department Division II, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guangli Han
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Orthodontic Department Division II, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Nabhan AN, Webster JD, Adams JJ, Blazer L, Everrett C, Eidenschenk C, Arlantico A, Fleming I, Brightbill HD, Wolters PJ, Modrusan Z, Seshagiri S, Angers S, Sidhu SS, Newton K, Arron JR, Dixit VM. Targeted alveolar regeneration with Frizzled-specific agonists. Cell 2023; 186:2995-3012.e15. [PMID: 37321220 DOI: 10.1016/j.cell.2023.05.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 03/24/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023]
Abstract
Wnt ligands oligomerize Frizzled (Fzd) and Lrp5/6 receptors to control the specification and activity of stem cells in many species. How Wnt signaling is selectively activated in different stem cell populations, often within the same organ, is not understood. In lung alveoli, we show that distinct Wnt receptors are expressed by epithelial (Fzd5/6), endothelial (Fzd4), and stromal (Fzd1) cells. Fzd5 is uniquely required for alveolar epithelial stem cell activity, whereas fibroblasts utilize distinct Fzd receptors. Using an expanded repertoire of Fzd-Lrp agonists, we could activate canonical Wnt signaling in alveolar epithelial stem cells via either Fzd5 or, unexpectedly, non-canonical Fzd6. A Fzd5 agonist (Fzd5ag) or Fzd6ag stimulated alveolar epithelial stem cell activity and promoted survival in mice after lung injury, but only Fzd6ag promoted an alveolar fate in airway-derived progenitors. Therefore, we identify a potential strategy for promoting regeneration without exacerbating fibrosis during lung injury.
Collapse
Affiliation(s)
- Ahmad N Nabhan
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Joshua D Webster
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jarret J Adams
- AntlerA Therapeutics, 348 Hatch Drive, Foster City, CA 94404, USA
| | - Levi Blazer
- AntlerA Therapeutics, 348 Hatch Drive, Foster City, CA 94404, USA
| | - Christine Everrett
- Department of Molecular Discovery and Cancer Cell Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Celine Eidenschenk
- Department of Molecular Discovery and Cancer Cell Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alexander Arlantico
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Isabel Fleming
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hans D Brightbill
- Department of Translational Immunology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Paul J Wolters
- Department of Medicine, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Zora Modrusan
- Department of Microchemistry, Proteomics, Lipidomics and Next Generation Sequencing, Genentech, South San Francisco, CA 94080, USA
| | | | - Stephane Angers
- AntlerA Therapeutics, 348 Hatch Drive, Foster City, CA 94404, USA; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 1A2, Canada; Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sachdev S Sidhu
- AntlerA Therapeutics, 348 Hatch Drive, Foster City, CA 94404, USA; School of Pharmacy, University of Waterloo, Kitchener, ON N2G 1C5, Canada
| | - Kim Newton
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Joseph R Arron
- Department of Immunology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Vishva M Dixit
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
22
|
Shah R, Amador C, Chun ST, Ghiam S, Saghizadeh M, Kramerov AA, Ljubimov AV. Non-canonical Wnt signaling in the eye. Prog Retin Eye Res 2023; 95:101149. [PMID: 36443219 PMCID: PMC10209355 DOI: 10.1016/j.preteyeres.2022.101149] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/12/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022]
Abstract
Wnt signaling comprises a group of complex signal transduction pathways that play critical roles in cell proliferation, differentiation, and apoptosis during development, as well as in stem cell maintenance and adult tissue homeostasis. Wnt pathways are classified into two major groups, canonical (β-catenin-dependent) or non-canonical (β-catenin-independent). Most previous studies in the eye have focused on canonical Wnt signaling, and the role of non-canonical signaling remains poorly understood. Additionally, the crosstalk between canonical and non-canonical Wnt signaling in the eye has hardly been explored. In this review, we present an overview of available data on ocular non-canonical Wnt signaling, including developmental and functional aspects in different eye compartments. We also discuss important changes of this signaling in various ocular conditions, such as keratoconus, aniridia-related keratopathy, diabetes, age-related macular degeneration, optic nerve damage, pathological angiogenesis, and abnormalities in the trabecular meshwork and conjunctival cells, and limbal stem cell deficiency.
Collapse
Affiliation(s)
- Ruchi Shah
- Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Regenerative Medicine Institute Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cynthia Amador
- Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Regenerative Medicine Institute Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steven T Chun
- Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Regenerative Medicine Institute Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA; University of California Los Angeles, Los Angeles, CA, USA
| | - Sean Ghiam
- Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Regenerative Medicine Institute Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Sackler School of Medicine, New York State/American Program of Tel Aviv University, Tel Aviv, Israel
| | - Mehrnoosh Saghizadeh
- Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Regenerative Medicine Institute Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA; David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Andrei A Kramerov
- Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Regenerative Medicine Institute Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexander V Ljubimov
- Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Regenerative Medicine Institute Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA; David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Klumpe HE, Garcia-Ojalvo J, Elowitz MB, Antebi YE. The computational capabilities of many-to-many protein interaction networks. Cell Syst 2023; 14:430-446. [PMID: 37348461 PMCID: PMC10318606 DOI: 10.1016/j.cels.2023.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/14/2023] [Accepted: 05/11/2023] [Indexed: 06/24/2023]
Abstract
Many biological circuits comprise sets of protein variants that interact with one another in a many-to-many, or promiscuous, fashion. These architectures can provide powerful computational capabilities that are especially critical in multicellular organisms. Understanding the principles of biochemical computations in these circuits could allow more precise control of cellular behaviors. However, these systems are inherently difficult to analyze, due to their large number of interacting molecular components, partial redundancies, and cell context dependence. Here, we discuss recent experimental and theoretical advances that are beginning to reveal how promiscuous circuits compute, what roles those computations play in natural biological contexts, and how promiscuous architectures can be applied for the design of synthetic multicellular behaviors.
Collapse
Affiliation(s)
- Heidi E Klumpe
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA
| | - Jordi Garcia-Ojalvo
- Department of Medicine and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain.
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Yaron E Antebi
- Department of Molecular Genetics, Weizmann Institute of Science 76100, Rehovot, Israel.
| |
Collapse
|
24
|
Carr HS, Zuo Y, Frost JA. The Wnt pathway protein Dvl1 targets somatostatin receptor 2 for lysosome-dependent degradation. J Biol Chem 2023; 299:104645. [PMID: 36965619 PMCID: PMC10164914 DOI: 10.1016/j.jbc.2023.104645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/27/2023] Open
Abstract
The Somatostatin receptor 2 (Sstr2) is a heterotrimeric G protein-coupled receptor that is highly expressed in neuroendocrine tumors and is a common pharmacological target for intervention. Unfortunately, not all neuroendocrine tumors express Sstr2, and Sstr2 expression can be downregulated with prolonged agonist use. Sstr2 is rapidly internalized following agonist stimulation and, in the short term, is quantitatively recycled back to the plasma membrane. However, mechanisms controlling steady state expression of Sstr2 in the absence of agonist are less well described. Here, we show that Sstr2 interacts with the Wnt pathway protein Dvl1 in a ligand-independent manner to target Sstr2 for lysosomal degradation. Interaction of Sstr2 with Dvl1 does not affect receptor internalization, recycling, or signaling to adenylyl cyclase but does suppress agonist-stimulated ERK1/2 activation. Importantly, Dvl1-dependent degradation of Sstr2 can be stimulated by overexpression of Wnts and treatment of cells with Wnt pathway inhibitors can boost Sstr2 expression in neuroendocrine tumor cells. Taken together, this study identifies for the first time a mechanism that targets Sstr2 for lysosomal degradation that is independent of Sstr2 agonist and can be potentiated by Wnt ligand. Intervention in this signaling mechanism has the potential to elevate Sstr2 expression in neuroendocrine tumors and enhance Sstr2-directed therapies.
Collapse
Affiliation(s)
- Heather S Carr
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, USA
| | - Yan Zuo
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, USA
| | - Jeffrey A Frost
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, USA.
| |
Collapse
|
25
|
Common and selective signal transduction mechanisms of GPCRs. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 195:89-100. [PMID: 36707157 DOI: 10.1016/bs.pmbts.2022.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
G protein-coupled receptors (GPCRs) are coupled by four major subfamilies of G proteins. GPCR coupling is processed through a combination of common and selective activation mechanisms together. Common mechanisms are shared for a group of receptors. Recently, researchers managed to identify shared activation pathways for the GPCRs belonging to the same subfamilies. On the other hand, selective mechanisms are responsible for the variations within activation mechanisms. Selective processes can regulate subfamily-specific interactions between the receptor and the G proteins, and intermediate receptor conformations are required to couple particular G proteins through G protein-specific activation mechanisms. Moreover, G proteins can also selectively interact with RGS (regulators of G protein signaling) proteins as well. Selective processes modulate the signaling profile of the receptor and the tissue they are present. This chapter summarizes the recent research conducted on common and selective signal transduction mechanisms within GPCRs from an evolutionary perspective.
Collapse
|
26
|
Ge Q, Teng M, Li X, Guo Q, Tao Y. An epitope-directed selection strategy facilitating the identification of Frizzled receptor selective antibodies. Structure 2023; 31:33-43.e5. [PMID: 36513066 DOI: 10.1016/j.str.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/27/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022]
Abstract
The lack of incorporating epitope information into the selection process makes the conventional antibody screening method less effective in identifying antibodies with desired functions. Here, we developed an epitope-directed antibody selection method by designing a directed library favoring the target epitope and a precise "counter" antigen for clearing irrelevant binders in the library. With this method, we successfully isolated an antibody, pF7_A5, that targets the less conserved region on the FZD2/7 CRD as designed. Guided by the structure of pF7_A5-FZD2CRD, a further round of evolution was conducted together with the "counter" antigen selection strategy, and ultimately, an FZD2-specific antibody and an FZD7-preferred antibody were obtained. Because of targeting the predefined functional site, all these antibodies exhibited the expected modulatory activity on the Wnt pathway. Together, the method developed here will be useful in antibody drug discovery, and the identified FZD antibodies will have clinical potential in FZD-related cancer therapy.
Collapse
Affiliation(s)
- Qiangqiang Ge
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China
| | - Maikun Teng
- Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Xu Li
- Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Qiong Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China.
| | - Yuyong Tao
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Ministry of Education Key Laboratory for Membraneless Organelles & Cellular Dynamics, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027 Hefei, P.R. China; Joint Laboratory of Innovation in Life Sciences University of Science and Technology of China (USTC) and Changchun Zhuoyi Biological Co. Ltd., 130616 Changchun, P.R. China.
| |
Collapse
|
27
|
Wnt signaling and the regulation of pluripotency. Curr Top Dev Biol 2023; 153:95-119. [PMID: 36967203 DOI: 10.1016/bs.ctdb.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The role of Wnt signaling in stem cells has been mired in seemingly contradictory findings. On one hand, Wnt has been heralded as a self-renewal factor. On the other hand, Wnt's association with differentiation and lineage commitment is indisputable. This apparent contradiction is particularly evident in pluripotent stem cells, where Wnt promotes self-renewal as well as differentiation. To resolve this discrepancy one must delve into fundamental principles of pluripotency and gain an appreciation for the concept of pluripotency states, which exist in a continuum with intermediate metastable states, some of which have been stabilized in vitro. Wnt signaling is a critical regulator of transitions between pluripotent states. Here, we will discuss Wnt's roles in maintaining pluripotency, promoting differentiation, as well as stimulating reprogramming of somatic cells to an induced pluripotent state.
Collapse
|
28
|
Mani N, Nygaard R, Mancia F. Gone with the Wnt(less): a mechanistic perspective on the journey of Wnt. Biochem Soc Trans 2022; 50:1763-1772. [PMID: 36416660 PMCID: PMC11962534 DOI: 10.1042/bst20220634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022]
Abstract
Wnts are short-range signaling proteins, expressed in all metazoans from sponges to humans, critical for cell development and fate. There are 19 different Wnts in the human genome with varying expression levels and patterns, and post-translational modifications. Common to essentially all Wnts is the palmitoleation of a conserved serine by the O-acyltransferase PORCN in the endoplasmic reticulum (ER). All lipidated Wnts then bind a dedicated carrier Wntless (WLS), endowed with the task of transporting them from the ER to the plasma membrane, and ultimately facilitating their release to receptors on the Wnt-receiving cell to initiate signaling. Here, we will focus on the WLS-mediated transport step. There are currently two published structures, both obtained by single-particle cryo-electron microscopy of the Wnt/WLS complex: human Wnt8A-bound and human Wnt3A-bound WLS. We analyze the two Wnt/WLS structures - remarkably similar despite the sequence similarity between Wnt8A and Wnt3A being only ∼39% - to begin to understand the conserved nature of this binding mechanism, and ultimately how one carrier can accommodate a family of 19 different Wnts. By comparing how Wnt associates with WLS with how it binds to PORCN and FZD receptors, we can begin to speculate on mechanisms of Wnt transfer from PORCN to WLS, and from WLS to FZD, thus providing molecular-level insight into these essential steps of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Neha Mani
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rie Nygaard
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
29
|
Habib SJ, Acebrón SP. Wnt signalling in cell division: from mechanisms to tissue engineering. Trends Cell Biol 2022; 32:1035-1048. [PMID: 35717422 DOI: 10.1016/j.tcb.2022.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 01/21/2023]
Abstract
Wnt signalling is an essential player in tissue formation, notably in the regulation of stem cell function. Wnt signalling is best known for its roles in G1/S progression. However, a complex Wnt programme that also mediates mitotic progression and asymmetric cell division (ACD) is emerging. Recent developments in this area have provided mechanistic insights as well as tools to engineer or target Wnt signalling for translational and therapeutic purposes. Here, we discuss the bidirectional relationship between Wnt activity and mitosis. We emphasise how various Wnt-dependent mechanisms control spindle dynamics, chromosome segregation, and ACD. Finally, we illustrate how knowledge about these mechanisms has been successfully employed in tissue engineering for regenerative medicine applications.
Collapse
Affiliation(s)
- Shukry J Habib
- Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7a, CH-1005 Lausanne, Switzerland.
| | - Sergio P Acebrón
- Centre for Organismal Studies (COS), University of Heidelberg, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany.
| |
Collapse
|
30
|
Liburkin-Dan T, Nir-Zvi I, Razon H, Kessler O, Neufeld G. Knock-Out of the Five Lysyl-Oxidase Family Genes Enables Identification of Lysyl-Oxidase Pro-Enzyme Regulated Genes. Int J Mol Sci 2022; 23:ijms231911322. [PMID: 36232621 PMCID: PMC9570307 DOI: 10.3390/ijms231911322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/12/2022] [Accepted: 09/17/2022] [Indexed: 12/02/2022] Open
Abstract
The five lysyl-oxidase genes share similar enzymatic activities and contribute to tumor progression. We have knocked out the five lysyl-oxidase genes in MDA-MB-231 breast cancer cells using CRISPR/Cas9 in order to identify genes that are regulated by LOX but not by other lysyl-oxidases and in order to study such genes in more mechanistic detail in the future. Re-expression of the full-length cDNA encoding LOX identified four genes whose expression was downregulated in the knock-out cells and rescued following LOX re-expression but not re-expression of other lysyl-oxidases. These were the AGR2, STOX2, DNAJB11 and DNAJC3 genes. AGR2 and STOX2 were previously identified as promoters of tumor progression. In addition, we identified several genes that were not downregulated in the knock-out cells but were strongly upregulated following LOX or LOXL3 re-expression. Some of these, such as the DERL3 gene, also promote tumor progression. There was very little proteolytic processing of the re-expressed LOX pro-enzyme in the MDA-MB-231 cells, while in the HEK293 cells, the LOX pro-enzyme was efficiently cleaved. We introduced point mutations into the known BMP-1 and ADAMTS2/14 cleavage sites of LOX. The BMP-1 mutant was secreted but not cleaved, while the LOX double mutant dmutLOX was not cleaved or secreted. However, even in the presence of the irreversible LOX inhibitor β-aminoproprionitrile (BAPN), these point-mutated LOX variants induced the expression of these genes, suggesting that the LOX pro-enzyme has hitherto unrecognized biological functions.
Collapse
|
31
|
Mahoney JP, Bruguera ES, Vasishtha M, Killingsworth LB, Kyaw S, Weis WI. PI(4,5)P 2-stimulated positive feedback drives the recruitment of Dishevelled to Frizzled in Wnt-β-catenin signaling. Sci Signal 2022; 15:eabo2820. [PMID: 35998232 PMCID: PMC9528458 DOI: 10.1126/scisignal.abo2820] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the Wnt-β-catenin pathway, Wnt binding to Frizzled (Fzd) and LRP5 or LRP6 (LRP5/6) co-receptors inhibits the degradation of the transcriptional coactivator β-catenin by recruiting the cytosolic effector Dishevelled (Dvl). Polymerization of Dvl at the plasma membrane recruits the β-catenin destruction complex, enabling the phosphorylation of LRP5/6, a key step in inhibiting β-catenin degradation. Using purified Fzd proteins reconstituted in lipid nanodiscs, we investigated the factors that promote the recruitment of Dvl to the plasma membrane. We found that the affinity of Fzd for Dvl was not affected by Wnt ligands, in contrast to other members of the GPCR superfamily for which the binding of extracellular ligands affects the affinity for downstream transducers. Instead, Fzd-Dvl binding was enhanced by increased concentration of the lipid PI(4,5)P2, which is generated by Dvl-associated lipid kinases in response to Wnt and which is required for LRP5/6 phosphorylation. Moreover, binding to Fzd did not promote Dvl DEP domain dimerization, which has been proposed to be required for signaling downstream of Fzd. Our findings suggest a positive feedback loop in which Wnt-stimulated local PI(4,5)P2 production enhances Dvl recruitment and further PI(4,5)P2 production to support Dvl polymerization, LRP5/6 phosphorylation, and β-catenin stabilization.
Collapse
Affiliation(s)
- Jacob P Mahoney
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Elise S Bruguera
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Mansi Vasishtha
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Lauren B Killingsworth
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Saw Kyaw
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - William I Weis
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| |
Collapse
|
32
|
Superresolution microscopy localizes endogenous Dvl2 to Wnt signaling-responsive biomolecular condensates. Proc Natl Acad Sci U S A 2022; 119:e2122476119. [PMID: 35867833 PMCID: PMC9335300 DOI: 10.1073/pnas.2122476119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Wnt signaling governs cell fate and tissue polarity across species. The Dishevelled proteins are central to Wnt signaling cascades. Wnt-mediated multiprotein complexes such as the “signalosome” and the “destruction complex” have been proposed to represent biomolecular condensates. These nonmembranous, specialized compartments have been suggested to form through liquid–liquid phase separation and ensure correctly proceeding physiological reactions. Although biomolecular condensates have increasingly been studied, key questions remain regarding, for example, their architecture and physiological regulation. Here, superresolution microscopy after endogenous labeling of Dishevelled-2 gives insights into protein functions and Wnt signaling at physiological levels. It reveals the distinct molecular architecture of endogenous Wnt condensates at single-molecule resolution and illustrates close interactions at the centrosome. During organismal development, homeostasis, and disease, Dishevelled (Dvl) proteins act as key signaling factors in beta-catenin–dependent and beta-catenin–independent Wnt pathways. While their importance for signal transmission has been genetically demonstrated in many organisms, our mechanistic understanding is still limited. Previous studies using overexpressed proteins showed Dvl localization to large, punctate-like cytoplasmic structures that are dependent on its DIX domain. To study Dvl’s role in Wnt signaling, we genome engineered an endogenously expressed Dvl2 protein tagged with an mEos3.2 fluorescent protein for superresolution imaging. First, we demonstrate the functionality and specificity of the fusion protein in beta-catenin–dependent and beta-catenin–independent signaling using multiple independent assays. We performed live-cell imaging of Dvl2 to analyze the dynamic formation of the supramolecular cytoplasmic Dvl2_mEos3.2 condensates. While overexpression of Dvl2_mEos3.2 mimics the previously reported formation of abundant large “puncta,” supramolecular condensate formation at physiological protein levels is only observed in a subset of cells with approximately one per cell. We show that, in these condensates, Dvl2 colocalizes with Wnt pathway components at gamma-tubulin and CEP164-positive centrosomal structures and that the localization of Dvl2 to these condensates is Wnt dependent. Single-molecule localization microscopy using photoactivated localization microscopy (PALM) of mEos3.2 in combination with DNA-PAINT demonstrates the organization and repetitive patterns of these condensates in a cell cycle–dependent manner. Our results indicate that the localization of Dvl2 in supramolecular condensates is coordinated dynamically and dependent on cell state and Wnt signaling levels. Our study highlights the formation of endogenous and physiologically regulated biomolecular condensates in the Wnt pathways at single-molecule resolution.
Collapse
|
33
|
Hussain B, Fang C, Huang X, Feng Z, Yao Y, Wang Y, Chang J. Endothelial β-Catenin Deficiency Causes Blood-Brain Barrier Breakdown via Enhancing the Paracellular and Transcellular Permeability. Front Mol Neurosci 2022; 15:895429. [PMID: 35615065 PMCID: PMC9125181 DOI: 10.3389/fnmol.2022.895429] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/04/2022] [Indexed: 01/24/2023] Open
Abstract
Disruption of the blood-brain barrier (BBB) causes or contributes to neuronal dysfunction and several central nervous system (CNS) disorders. Wnt/β-catenin signaling is essential for maintaining the integrity of the adult BBB in physiological and pathological conditions, including stroke. However, how the impairment of the endothelial Wnt/β-catenin signaling results in BBB breakdown remains unclear. Furthermore, the individual contributions of different BBB permeability-inducing mechanisms, including intercellular junction damage, endothelial transcytosis, and fenestration, remains unexplored. Here, we induced β-catenin endothelial-specific conditional knockout (ECKO) in adult mice and determined its impact on BBB permeability and the underlying mechanism. β-catenin ECKO reduced the levels of active β-catenin and the mRNA levels of Wnt target genes in mice, indicating downregulation of endothelial Wnt/β-catenin signaling. β-catenin ECKO mice displayed severe and widespread leakage of plasma IgG and albumin into the cerebral cortex, which was absent in wild-type controls. Mechanistically, both the paracellular and transcellular transport routes were disrupted in β-catenin ECKO mice. First, β-catenin ECKO reduced the tight junction protein levels and disrupted the intercellular junction ultrastructure in the brain endothelium. Second, β-catenin ECKO substantially increased the number of endothelial vesicles and caveolae-mediated transcytosis through downregulating Mfsd2a and upregulating caveolin-1 expression. Interestingly, fenestration and upregulated expression of the fenestration marker Plvap were not observed in β-catenin ECKO mice. Overall, our study reveals that endothelial Wnt/β-catenin signaling maintains adult BBB integrity via regulating the paracellular as well as transcellular permeability. These findings may have broad applications in understanding and treatment of CNS disorders involving BBB disruption.
Collapse
Affiliation(s)
- Basharat Hussain
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Fang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaowen Huang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ziying Feng
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuxuan Yao
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
34
|
De Re V, Alessandrini L, Brisotto G, Caggiari L, De Zorzi M, Casarotto M, Miolo G, Puglisi F, Garattini SK, Lonardi S, Cannizzaro R, Canzonieri V, Fassan M, Steffan A. HER2-CDH1 Interaction via Wnt/B-Catenin Is Associated with Patients' Survival in HER2-Positive Metastatic Gastric Adenocarcinoma. Cancers (Basel) 2022; 14:1266. [PMID: 35267574 PMCID: PMC8909509 DOI: 10.3390/cancers14051266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
Trastuzumab is a human epidermal growth factor receptor 2 (HER2) inhibitor used to treat HER2+ metastatic gastric cancer (mGC). The present study aims to investigate the relationship between CDH1 mRNA expression and HER2-positivity in mGC using a multiplexed gene expression profile in two series of gastric cancer (GC): Series 1 (n = 38): HER2+ and HER2- mGC; Series 2 (n = 36) HER2- GC with and without metastasis. To confirm the results, the same expression profiles were analyzed in 354 GC from The Cancer Genome Atlas (TCGA) dataset. The difference in gene expression connected HER2 overexpression with canonical wingless-type (Wnt)/β-catenin pathway and immunohistochemical (IHC) expression loss of E-cadherin (E-CAD). CDH1 mRNA expression was simultaneously associated with the rs16260-A variant and an increase in E-CAD expression. Differences in retinoic acid receptor alfa (RARA), RPL19 (coding for the 60S ribosomal L19 protein), catenin delta 1 (CTNND1), and epidermal growth factor (EGF) mRNA levels-all included in the Wnt/β-catenin pathway-were found associated with overall survival (OS). RARA, CTNND1, and EGF resulted in independent OS prognostic factors. EGF was confirmed as an independent factor along with TNM stage in HER2-overpressed mGC from TCGA collection. Our study highlighted factors involved in the WNT/β-catenin pathway that interconnected E-CAD with HER2 overexpression and patient survival.
Collapse
Affiliation(s)
- Valli De Re
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (G.B.); (L.C.); (M.D.Z.); (M.C.); (A.S.)
| | - Lara Alessandrini
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy; (L.A.); (M.F.)
| | - Giulia Brisotto
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (G.B.); (L.C.); (M.D.Z.); (M.C.); (A.S.)
| | - Laura Caggiari
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (G.B.); (L.C.); (M.D.Z.); (M.C.); (A.S.)
| | - Mariangela De Zorzi
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (G.B.); (L.C.); (M.D.Z.); (M.C.); (A.S.)
| | - Mariateresa Casarotto
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (G.B.); (L.C.); (M.D.Z.); (M.C.); (A.S.)
| | - Gianmaria Miolo
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (G.M.); (F.P.)
| | - Fabio Puglisi
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (G.M.); (F.P.)
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | | | - Sara Lonardi
- Oncology Unit 3, Veneto Institute of Oncology, Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS), 35128 Padova, Italy;
| | - Renato Cannizzaro
- Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Matteo Fassan
- Surgical Pathology and Cytopathology Unit, Department of Medicine (DIMED), University of Padova, 35128 Padova, Italy; (L.A.); (M.F.)
- Oncology Unit 3, Veneto Institute of Oncology, Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS), 35128 Padova, Italy;
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (G.B.); (L.C.); (M.D.Z.); (M.C.); (A.S.)
| |
Collapse
|
35
|
Abstract
The Wnt pathway is central to a host of developmental and disease-related processes. The remarkable conservation of this intercellular signaling cascade throughout metazoan lineages indicates that it coevolved with multicellularity to regulate the generation and spatial arrangement of distinct cell types. By regulating cell fate specification, mitotic activity, and cell polarity, Wnt signaling orchestrates development and tissue homeostasis, and its dysregulation is implicated in developmental defects, cancer, and degenerative disorders. We review advances in our understanding of this key pathway, from Wnt protein production and secretion to relay of the signal in the cytoplasm of the receiving cell. We discuss the evolutionary history of this pathway as well as endogenous and synthetic modulators of its activity. Finally, we highlight remaining gaps in our knowledge of Wnt signal transduction and avenues for future research. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Ellen Youngsoo Rim
- Howard Hughes Medical Institute, Department of Developmental Biology, and Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California, USA;
| | - Hans Clevers
- Hubrecht Institute and Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Utrecht, The Netherlands
| | - Roel Nusse
- Howard Hughes Medical Institute, Department of Developmental Biology, and Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California, USA;
| |
Collapse
|
36
|
Signalling dynamics in embryonic development. Biochem J 2021; 478:4045-4070. [PMID: 34871368 PMCID: PMC8718268 DOI: 10.1042/bcj20210043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 02/08/2023]
Abstract
In multicellular organisms, cellular behaviour is tightly regulated to allow proper embryonic development and maintenance of adult tissue. A critical component in this control is the communication between cells via signalling pathways, as errors in intercellular communication can induce developmental defects or diseases such as cancer. It has become clear over the last years that signalling is not static but varies in activity over time. Feedback mechanisms present in every signalling pathway lead to diverse dynamic phenotypes, such as transient activation, signal ramping or oscillations, occurring in a cell type- and stage-dependent manner. In cells, such dynamics can exert various functions that allow organisms to develop in a robust and reproducible way. Here, we focus on Erk, Wnt and Notch signalling pathways, which are dynamic in several tissue types and organisms, including the periodic segmentation of vertebrate embryos, and are often dysregulated in cancer. We will discuss how biochemical processes influence their dynamics and how these impact on cellular behaviour within multicellular systems.
Collapse
|
37
|
Dai R, Wu M, Zhang Y, Zhu Z, Shi J. G protein nucleolar 3 promotes Non-Hodgkin lymphoma progression by activating the Wnt/β-catenin signaling pathway. Exp Cell Res 2021; 409:112911. [PMID: 34762898 DOI: 10.1016/j.yexcr.2021.112911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/18/2021] [Accepted: 11/06/2021] [Indexed: 10/19/2022]
Abstract
G protein nucleolar 3 (GNL3), which acts as an oncoprotein in various carcinomas, is associated with tumor progression; however, little is known regarding GNL3 function in non-Hodgkin lymphoma (NHL). In this study, we first used in silico analysis to determine associations between GNL3 and diffuse large B-cell lymphoma (DLBCL). We then examined the effect of GNL3 on NHL progression, including cell proliferation, apoptosis, and cell cycle progression, and determined its underlying molecular mechanism using in vitro lymphoma cell lines and in vivo mouse xenograft models. We found that GNL3 mRNA levels were markedly higher in DLBCL tissues than in normal tissues, with these higher levels associated with poor prognosis. Additionally, GNL3 overexpression promoted NHL cell proliferation and cell cycle progression and reduced apoptosis in vitro, and enhanced tumorigenesis in an in vivo xenograft model. Moreover, we found that GNL3 upregulated the levels of Wnt/β-catenin signaling pathway-related factors and downstream target genes, whereas the opposite result was observed in GNL3-silenced cells. Furthermore, a rescue experiment using a Wnt/β-catenin inhibitor (XAV939) confirmed that GNL3 promotes NHL progression by activating the Wnt/β-catenin signaling pathway. These findings demonstrated that GNL3 functions as an oncogenic driver in NHL via the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Rongqin Dai
- Department of Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan province, PR China
| | - Meirong Wu
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian province, PR China
| | - Yin Zhang
- Department of Hematology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan province, PR China
| | - Zunmin Zhu
- Department of Hematology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan province, PR China.
| | - Jie Shi
- Department of Hematology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan province, PR China.
| |
Collapse
|
38
|
Zougros A, Michelli M, Chatziandreou I, Nonni A, Gakiopoulou H, Michalopoulos NV, Lazaris AC, Saetta AA. mRNA coexpression patterns of Wnt pathway components and their clinicopathological associations in breast and colorectal cancer. Pathol Res Pract 2021; 227:153649. [PMID: 34656913 DOI: 10.1016/j.prp.2021.153649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 11/24/2022]
Abstract
Aberrant Wnt signaling is implicated in carcinogenesis triggering efforts for the development of new therapeutic agents, many of which have entered clinical trials. We extend our previous analysis of WNT3, FZD7, LEF1 expression levels in breast and colorectal cancer including WNT2, FZD4 and β-catenin expression, in an effort to delineate their relative expression levels along with concurrent expression patterns and possible prognostic value. We analyzed 82 breast and 102 colorectal carcinomas for relative mRNA expression levels of the investigated genes by RT-PCR relative quantification with the ΔΔCt method. Statistical analysis was performed in order to determine associations of relative mRNA expression and linear correlations. β-catenin expression was determined by immunochemistry. Regarding breast carcinomas, decreased relative mRNA expression levels of WNT2, FZD4 were found frequently and WNT2 expression was correlated with ER/ PR status (p = 0.045/p = 0.028), whereas β-catenin with grade (p = 0.026). In colorectal carcinomas, increased relative mRNA expression levels of WNT2 and FZD4 were found in 59% and 32% of cases respectively, whereas β-catenin showed decreased mRNA expression levels in 57% of cases and a correlation with pN-category (p = 0.037). Linear correlations were observed between WNT2/FZD4 (R=0.542, p < 0.001), WNT2/β-catenin (R=0.254, p = 0.010), FZD4/β-catenin (R=0.406, p < 0.001) expression and a correlation between mRNA expression and membranous/cytoplasmic β-catenin emerged (p = 0.039/0.046). Our results suggest a possible clinical significance for Wnt pathway gene expression levels in both tumour types. The concurrent expression of the investigated genes as well as the different expression profiles, underlines the complexity of this pathway and the necessity of patient selection in order to maximize the efficacy of drugs targeting Wnt pathway.
Collapse
Affiliation(s)
- Alexandros Zougros
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, Athens, Greece
| | - Maria Michelli
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, Athens, Greece
| | - Ilenia Chatziandreou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, Athens, Greece
| | - Afroditi Nonni
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, Athens, Greece
| | - Harikleia Gakiopoulou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, Athens, Greece
| | - Nikolaos V Michalopoulos
- Fourth Department of Surgery, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Rimini 1, Haidari, Athens, Greece
| | - Andreas C Lazaris
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, Athens, Greece
| | - Angelica A Saetta
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, Athens, Greece.
| |
Collapse
|
39
|
Sharma A, Mir R, Galande S. Epigenetic Regulation of the Wnt/β-Catenin Signaling Pathway in Cancer. Front Genet 2021; 12:681053. [PMID: 34552611 PMCID: PMC8450413 DOI: 10.3389/fgene.2021.681053] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Studies over the past four decades have elucidated the role of Wnt/β-catenin mediated regulation in cell proliferation, differentiation and migration. These processes are fundamental to embryonic development, regeneration potential of tissues, as well as cancer initiation and progression. In this review, we focus on the epigenetic players which influence the Wnt/β-catenin pathway via modulation of its components and coordinated regulation of the Wnt target genes. The role played by crosstalk with other signaling pathways mediating tumorigenesis is also elaborated. The Hippo/YAP pathway is particularly emphasized due to its extensive crosstalk via the Wnt destruction complex. Further, we highlight the recent advances in developing potential therapeutic interventions targeting the epigenetic machinery based on the characterization of these regulatory networks for effective treatment of various cancers and also for regenerative therapies.
Collapse
Affiliation(s)
- Ankita Sharma
- Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Rafeeq Mir
- Centre for Interdisciplinary Research and Innovations, University of Kashmir, Srinagar, India
| | - Sanjeev Galande
- Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research, Pune, India.,Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
| |
Collapse
|
40
|
Na L, Wang Z, Bai Y, Sun Y, Dong D, Wang W, Zhao C. WNT7B represses epithelial-mesenchymal transition and stem-like properties in bladder urothelial carcinoma. Biochim Biophys Acta Mol Basis Dis 2021; 1868:166271. [PMID: 34562599 DOI: 10.1016/j.bbadis.2021.166271] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/01/2021] [Accepted: 09/15/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Recurrence and metastasis are the major problems of bladder urothelial carcinoma, which mainly attribute to tumor cell stemness, epithelial-mesenchymal transition (EMT) and chemoresistance. METHODS TCGA database was interrogated for gene mRNA expression in bladder urothelial carcinoma samples. CCLE database was interrogated for gene mRNA expression in bladder cancer cell lines. The correlation between two genes was analyzed by Pearson statistics. 37 human bladder urothelial carcinoma specimens were adopted for immunohistochemistry. Bladder cancer cells RT4, J82, and UM-UC-3 were used to carry out loss and gain of function studies. Kaplan-Meier method was performed to analyze the overall survival. FINDINGS WNT7B is downregulated in high-grade bladder urothelial carcinomas. Low WNT7B expression is associated with unfavorable prognosis. Loss and gain of function studies showed that WNT7B inhibits bladder urothelial carcinoma cell EMT, stem-like properties and chemoresistance. FZD5, a specific receptor for WNT7B, mediates WNT7B signaling. ELF3 is a downstream component of WNT7B signaling, which transcriptionally modulates NOTCH1, a tumor suppressor in bladder urothelial carcinoma. INTERPRETATION These data demonstrate that WNT7B/FZD5-ELF3-NOTCH1 signaling functions as a tumor-suppressing pathway in bladder urothelial carcinoma.
Collapse
Affiliation(s)
- Lei Na
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China; Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhuo Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yu Bai
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China; Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Dan Dong
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China.
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China.
| |
Collapse
|
41
|
Peng H, Zheng Y, Zhao Z, Li J. Multigene editing: current approaches and beyond. Brief Bioinform 2021; 22:bbaa396. [PMID: 33428725 DOI: 10.1093/bib/bbaa396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/15/2020] [Accepted: 12/03/2020] [Indexed: 11/14/2022] Open
Abstract
CRISPR/Cas9 multigene editing is an active and widely studied topic in the fields of biomedicine and biology. It involves a simultaneous participation of multiple single-guide RNAs (sgRNAs) to edit multiple target genes in a way that each gene is edited by one of these sgRNAs. There are possibly numerous sgRNA candidates capable of on-target editing on each of these genes with various efficiencies. Meanwhile, each of these sgRNA candidates may cause unwanted off-target editing at many other genes. Therefore, selection optimization of these multiple sgRNAs is demanded so as to minimize the number of sgRNAs and thus reduce the collective negative effects caused by the off-target editing. This survey reviews wet-laboratory approaches to the implementation of multigene editing and their needs of computational tools for better design. We found that though off-target editing is unavoidable during the gene editing, those disfavored cuttings by some target genes' sgRNAs can potentially become on-target editing sites for some other genes of interests. This off-to-on role conversion is beneficial to optimize the sgRNA selection in multigene editing. We present a preference cutting score to assess those beneficial off-target cutting sites, which have a few mismatches with their host genes' on-target editing sites. These potential sgRNAs can be prioritized for recommendation via ranking their on-target average cutting efficiency, the total off-target site number and their average preference cutting score. We also present case studies on cancer-associated genes to demonstrate tremendous usefulness of the new method.
Collapse
Affiliation(s)
- Hui Peng
- Data Science Institute, University of Technology Sydney, PO Box 123, Ultimo, NSW 2007, Australia
- School of Computing, National University of Singapore, 13 Computing Drive, 117417, Singapore
| | - Yi Zheng
- Data Science Institute, University of Technology Sydney, PO Box 123, Ultimo, NSW 2007, Australia
| | - Zhixun Zhao
- Data Science Institute, University of Technology Sydney, PO Box 123, Ultimo, NSW 2007, Australia
| | - Jinyan Li
- Data Science Institute, University of Technology Sydney, PO Box 123, Ultimo, NSW 2007, Australia
| |
Collapse
|
42
|
Koch S. Regulation of Wnt Signaling by FOX Transcription Factors in Cancer. Cancers (Basel) 2021; 13:cancers13143446. [PMID: 34298659 PMCID: PMC8307807 DOI: 10.3390/cancers13143446] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Cancer is caused by a breakdown of cell-to-cell communication, which results in the unrestricted expansion of cells within a tissue. In many cases, tumor growth is maintained by the continuous activation of cell signaling programs that normally drive embryonic development and wound repair. In this review article, I discuss how one of the largest human protein families, namely FOX proteins, controls the activity of the Wnt pathway, a major regulatory signaling cascade in developing organisms and adult stem cells. Evidence suggests that there is considerable crosstalk between FOX proteins and the Wnt pathway, which contributes to cancer initiation and progression. A better understanding of FOX biology may therefore lead to the development of new targeted treatments for many types of cancer. Abstract Aberrant activation of the oncogenic Wnt signaling pathway is a hallmark of numerous types of cancer. However, in many cases, it is unclear how a chronically high Wnt signaling tone is maintained in the absence of activating pathway mutations. Forkhead box (FOX) family transcription factors are key regulators of embryonic development and tissue homeostasis, and there is mounting evidence that they act in part by fine-tuning the Wnt signaling output in a tissue-specific and context-dependent manner. Here, I review the diverse ways in which FOX transcription factors interact with the Wnt pathway, and how the ectopic reactivation of FOX proteins may affect Wnt signaling activity in various types of cancer. Many FOX transcription factors are partially functionally redundant and exhibit a highly restricted expression pattern, especially in adults. Thus, precision targeting of individual FOX proteins may lead to safe treatment options for Wnt-dependent cancers.
Collapse
Affiliation(s)
- Stefan Koch
- Wallenberg Centre for Molecular Medicine (WCMM), Linköping University, 58185 Linköping, Sweden; ; Tel.: +46-132-829-69
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58185 Linköping, Sweden
| |
Collapse
|
43
|
Lu Y, Liu M, Yang J, Weissman SM, Pan X, Katz SG, Wang S. Spatial transcriptome profiling by MERFISH reveals fetal liver hematopoietic stem cell niche architecture. Cell Discov 2021; 7:47. [PMID: 34183665 PMCID: PMC8238952 DOI: 10.1038/s41421-021-00266-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/25/2021] [Indexed: 12/17/2022] Open
Abstract
The hematopoietic stem cell (HSC) niche has been extensively studied in bone marrow, yet a more systematic investigation into the microenvironment regulation of hematopoiesis in fetal liver is necessary. Here we investigate the spatial organization and transcriptional profile of individual cells in both wild type (WT) and Tet2−/− fetal livers, by multiplexed error robust fluorescence in situ hybridization. We find that specific pairs of fetal liver cell types are preferentially positioned next to each other. Ligand-receptor signaling molecule pairs such as Kitl and Kit are enriched in neighboring cell types. The majority of HSCs are in direct contact with endothelial cells (ECs) in both WT and Tet2−/− fetal livers. Loss of Tet2 increases the number of HSCs, and upregulates Wnt and Notch signaling genes in the HSC niche. Two subtypes of ECs, arterial ECs and sinusoidal ECs, and other cell types contribute distinct signaling molecules to the HSC niche. Collectively, this study provides a comprehensive picture and bioinformatic foundation for HSC spatial regulation in fetal liver.
Collapse
Affiliation(s)
- Yanfang Lu
- Department of Genetics, Yale School of Medicine, New Haven, USA.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, China
| | - Miao Liu
- Department of Genetics, Yale School of Medicine, New Haven, USA
| | - Jennifer Yang
- Department of Genetics, Yale School of Medicine, New Haven, USA
| | | | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China. .,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, China.
| | - Samuel G Katz
- Department of Pathology, Yale School of Medicine, New Haven, USA.
| | - Siyuan Wang
- Department of Genetics, Yale School of Medicine, New Haven, USA. .,Department of Cell Biology, Yale School of Medicine, New Haven, USA.
| |
Collapse
|
44
|
Ordonez LD, Melchor L, Greenow KR, Kendrick H, Tornillo G, Bradford J, Giles P, Smalley MJ. Reproductive history determines Erbb2 locus amplification, WNT signalling and tumour phenotype in a murine breast cancer model. Dis Model Mech 2021; 14:264801. [PMID: 34003256 PMCID: PMC8188886 DOI: 10.1242/dmm.048736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/25/2021] [Indexed: 11/20/2022] Open
Abstract
Understanding the mechanisms underlying tumour heterogeneity is key to the development of treatments that can target specific tumour subtypes. We have previously targeted CRE recombinase-dependent conditional deletion of the tumour suppressor genes Brca1, Brca2, p53 (also known as Trp53) and/or Pten to basal or luminal oestrogen receptor-negative (ER−) cells of the mouse mammary epithelium. We demonstrated that both the cell-of-origin and the tumour-initiating genetic lesions cooperate to influence mammary tumour phenotype. Here, we use a CRE-activated HER2 orthologue to specifically target HER2/ERBB2 oncogenic activity to basal or luminal ER− mammary epithelial cells and perform a detailed analysis of the tumours that develop. We find that, in contrast to our previous studies, basal epithelial cells are less sensitive to transformation by the activated NeuKI allele, with mammary epithelial tumour formation largely confined to luminal ER− cells. Histologically, most tumours that developed were classified as either adenocarcinomas of no special type or as metaplastic adenosquamous tumours. The former were typically characterized by amplification of the NeuNT/Erbb2 locus; in contrast, tumours displaying squamous metaplasia were enriched in animals that had been through at least one pregnancy and typically had lower levels of NeuNT/Erbb2 locus amplification but had activated canonical WNT signalling. Squamous changes in these tumours were associated with activation of the epidermal differentiation cluster. Thus, in this model of HER2 breast cancer, cell-of-origin, reproductive history, NeuNT/Erbb2 locus amplification and the activation of specific branches of the WNT signalling pathway all interact to drive inter-tumour heterogeneity. Summary: Using a mouse model of breast cancer, the authors show mammary epithelial cell-type sensitivity to transformation by HER2, as well as a change in tumour phenotype associated with reproductive history and driven by WNT signalling.
Collapse
Affiliation(s)
- Liliana D Ordonez
- European Cancer Stem Cell Research Institute and Cardiff School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Lorenzo Melchor
- European Cancer Stem Cell Research Institute and Cardiff School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Kirsty R Greenow
- European Cancer Stem Cell Research Institute and Cardiff School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Howard Kendrick
- European Cancer Stem Cell Research Institute and Cardiff School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | - Giusy Tornillo
- European Cancer Stem Cell Research Institute and Cardiff School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| | | | - Peter Giles
- Wales Gene Park, University Hospital Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Matthew J Smalley
- European Cancer Stem Cell Research Institute and Cardiff School of Biosciences, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| |
Collapse
|
45
|
Kozielewicz P, Shekhani R, Moser S, Bowin CF, Wesslowski J, Davidson G, Schulte G. Quantitative Profiling of WNT-3A Binding to All Human Frizzled Paralogues in HEK293 Cells by NanoBiT/BRET Assessments. ACS Pharmacol Transl Sci 2021; 4:1235-1245. [PMID: 34151213 PMCID: PMC8205236 DOI: 10.1021/acsptsci.1c00084] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Indexed: 02/06/2023]
Abstract
![]()
The WNT signaling
system governs critical processes during embryonic
development and tissue homeostasis, and its dysfunction can lead to
cancer. Details concerning selectivity and differences in relative
binding affinities of 19 mammalian WNTs to the cysteine-rich domain
(CRD) of their receptors—the ten mammalian Frizzleds (FZDs)—remain
unclear. Here, we used eGFP-tagged mouse WNT-3A for a systematic analysis
of WNT interaction with every human FZD paralogue in HEK293A cells.
Employing HiBiT-tagged full-length FZDs, we studied eGFP-WNT-3A binding
kinetics, saturation binding, and competition binding with commercially
available WNTs in live HEK293A cells using a NanoBiT/BRET-based assay.
Further, we generated receptor chimeras to dissect the contribution
of the transmembrane core to WNT-CRD binding. Our data pinpoint distinct
WNT-FZD selectivity and shed light on the complex WNT-FZD binding
mechanism. The methodological development described herein reveals
yet unappreciated details of the complexity of WNT signaling and WNT-FZD
interactions, providing further details with respect to WNT-FZD selectivity.
Collapse
Affiliation(s)
- Paweł Kozielewicz
- Section of Receptor Biology & Signaling, Dept. Physiology & Pharmacology, Karolinska Institutet, S-17165, Stockholm, Sweden
| | - Rawan Shekhani
- Section of Receptor Biology & Signaling, Dept. Physiology & Pharmacology, Karolinska Institutet, S-17165, Stockholm, Sweden
| | - Stefanie Moser
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), 76131, Karlsruhe, Germany
| | - Carl-Fredrik Bowin
- Section of Receptor Biology & Signaling, Dept. Physiology & Pharmacology, Karolinska Institutet, S-17165, Stockholm, Sweden
| | - Janine Wesslowski
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), 76131, Karlsruhe, Germany
| | - Gary Davidson
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), 76131, Karlsruhe, Germany
| | - Gunnar Schulte
- Section of Receptor Biology & Signaling, Dept. Physiology & Pharmacology, Karolinska Institutet, S-17165, Stockholm, Sweden
| |
Collapse
|
46
|
Landin Malt A, Clancy S, Hwang D, Liu A, Smith C, Smith M, Hatley M, Clemens C, Lu X. Non-Canonical Wnt Signaling Regulates Cochlear Outgrowth and Planar Cell Polarity via Gsk3β Inhibition. Front Cell Dev Biol 2021; 9:649830. [PMID: 33937247 PMCID: PMC8086559 DOI: 10.3389/fcell.2021.649830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/17/2021] [Indexed: 11/26/2022] Open
Abstract
During development, sensory hair cells (HCs) in the cochlea assemble a stereociliary hair bundle on their apical surface with planar polarized structure and orientation. We have recently identified a non-canonical, Wnt/G-protein/PI3K signaling pathway that promotes cochlear outgrowth and coordinates planar polarization of the HC apical cytoskeleton and alignment of HC orientation across the cochlear epithelium. Here, we determined the involvement of the kinase Gsk3β and the small GTPase Rac1 in non-canonical Wnt signaling and its regulation of the planar cell polarity (PCP) pathway in the cochlea. We provided the first in vivo evidence for Wnt regulation of Gsk3β activity via inhibitory Ser9 phosphorylation. Furthermore, we carried out genetic rescue experiments of cochlear defects caused by blocking Wnt secretion. We showed that cochlear outgrowth was partially rescued by genetic ablation of Gsk3β but not by expression of stabilized β-catenin; while PCP defects, including hair bundle polarity and junctional localization of the core PCP proteins Fzd6 and Dvl2, were partially rescued by either Gsk3β ablation or constitutive activation of Rac1. Our results identify Gsk3β and likely Rac1 as downstream components of non-canonical Wnt signaling and mediators of cochlear outgrowth, HC planar polarity, and localization of a subset of core PCP proteins in the cochlea.
Collapse
Affiliation(s)
- Andre Landin Malt
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| | - Shaylyn Clancy
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| | - Diane Hwang
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| | - Alice Liu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| | - Connor Smith
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| | - Margaret Smith
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| | - Maya Hatley
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| | - Christopher Clemens
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| | - Xiaowei Lu
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA, United States
| |
Collapse
|
47
|
Brunt L, Greicius G, Rogers S, Evans BD, Virshup DM, Wedgwood KCA, Scholpp S. Vangl2 promotes the formation of long cytonemes to enable distant Wnt/β-catenin signaling. Nat Commun 2021; 12:2058. [PMID: 33824332 PMCID: PMC8024337 DOI: 10.1038/s41467-021-22393-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/09/2021] [Indexed: 02/01/2023] Open
Abstract
Wnt signaling regulates cell proliferation and cell differentiation as well as migration and polarity during development. However, it is still unclear how the Wnt ligand distribution is precisely controlled to fulfil these functions. Here, we show that the planar cell polarity protein Vangl2 regulates the distribution of Wnt by cytonemes. In zebrafish epiblast cells, mouse intestinal telocytes and human gastric cancer cells, Vangl2 activation generates extremely long cytonemes, which branch and deliver Wnt protein to multiple cells. The Vangl2-activated cytonemes increase Wnt/β-catenin signaling in the surrounding cells. Concordantly, Vangl2 inhibition causes fewer and shorter cytonemes to be formed and reduces paracrine Wnt/β-catenin signaling. A mathematical model simulating these Vangl2 functions on cytonemes in zebrafish gastrulation predicts a shift of the signaling gradient, altered tissue patterning, and a loss of tissue domain sharpness. We confirmed these predictions during anteroposterior patterning in the zebrafish neural plate. In summary, we demonstrate that Vangl2 is fundamental to paracrine Wnt/β-catenin signaling by controlling cytoneme behaviour.
Collapse
Affiliation(s)
- Lucy Brunt
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Gediminas Greicius
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Sally Rogers
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Benjamin D Evans
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
- School of Psychological Science, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - David M Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Kyle C A Wedgwood
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
48
|
Giebel N, de Jaime-Soguero A, García Del Arco A, Landry JJM, Tietje M, Villacorta L, Benes V, Fernández-Sáiz V, Acebrón SP. USP42 protects ZNRF3/RNF43 from R-spondin-dependent clearance and inhibits Wnt signalling. EMBO Rep 2021; 22:e51415. [PMID: 33786993 PMCID: PMC8097334 DOI: 10.15252/embr.202051415] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/16/2021] [Accepted: 03/01/2021] [Indexed: 12/16/2022] Open
Abstract
The tumour suppressors RNF43 and ZNRF3 play a central role in development and tissue homeostasis by promoting the turnover of the Wnt receptors LRP6 and Frizzled (FZD). The stem cell growth factor R‐spondin induces auto‐ubiquitination and membrane clearance of ZNRF3/RNF43 to promote Wnt signalling. However, the deubiquitinase stabilising ZNRF3/RNF43 at the plasma membrane remains unknown. Here, we show that the USP42 antagonises R‐spondin by protecting ZNRF3/RNF43 from ubiquitin‐dependent clearance. USP42 binds to the Dishevelled interacting region (DIR) of ZNRF3 and stalls the R‐spondin‐LGR4‐ZNRF3 ternary complex by deubiquitinating ZNRF3. Accordingly, USP42 increases the turnover of LRP6 and Frizzled (FZD) receptors and inhibits Wnt signalling. Furthermore, we show that USP42 functions as a roadblock for paracrine Wnt signalling in colon cancer cells and mouse small intestinal organoids. We provide new mechanistic insights into the regulation R‐spondin and conclude that USP42 is crucial for ZNRF3/RNF43 stabilisation at the cell surface.
Collapse
Affiliation(s)
- Nicole Giebel
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | | | - Ana García Del Arco
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Jonathan J M Landry
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Marlene Tietje
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Laura Villacorta
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Vanesa Fernández-Sáiz
- Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Sergio P Acebrón
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
49
|
Shi W, Xu C, Gong Y, Wang J, Ren Q, Yan Z, Mei L, Tang C, Ji X, Hu X, Qv M, Hussain M, Zeng LH, Wu X. RhoA/Rock activation represents a new mechanism for inactivating Wnt/β-catenin signaling in the aging-associated bone loss. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:8. [PMID: 33655459 PMCID: PMC7925793 DOI: 10.1186/s13619-020-00071-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022]
Abstract
The Wnt/β-catenin signaling pathway appears to be particularly important for bone homeostasis, whereas nuclear accumulation of β-catenin requires the activation of Rac1, a member of the Rho small GTPase family. The aim of the present study was to investigate the role of RhoA/Rho kinase (Rock)-mediated Wnt/β-catenin signaling in the regulation of aging-associated bone loss. We find that Lrp5/6-dependent and Lrp5/6-independent RhoA/Rock activation by Wnt3a activates Jak1/2 to directly phosphorylate Gsk3β at Tyr216, resulting in Gsk3β activation and subsequent β-catenin destabilization. In line with these molecular events, RhoA loss- or gain-of-function in mouse embryonic limb bud ectoderms interacts genetically with Dkk1 gain-of-function to rescue the severe limb truncation phenotypes or to phenocopy the deletion of β-catenin, respectively. Likewise, RhoA loss-of-function in pre-osteoblasts robustly increases bone formation while gain-of-function decreases it. Importantly, high RhoA/Rock activity closely correlates with Jak and Gsk3β activities but inversely correlates with β-catenin signaling activity in bone marrow mesenchymal stromal cells from elderly male humans and mice, whereas systemic inhibition of Rock therefore activates the β-catenin signaling to antagonize aging-associated bone loss. Taken together, these results identify RhoA/Rock-dependent Gsk3β activation and subsequent β-catenin destabilization as a hitherto uncharacterized mechanism controlling limb outgrowth and bone homeostasis.
Collapse
Affiliation(s)
- Wei Shi
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
- Department of Biology and Genetics, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
- Department of Orthopeadic Surgery of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Ying Gong
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Jirong Wang
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Qianlei Ren
- Department of Pharmacology, Zhejiang University City College, 51 Huzhou Street, Hangzhou, 310015, China
| | - Ziyi Yan
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Liu Mei
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Chao Tang
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Xing Ji
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
- Translational Research Program in Pediatric Orthopaedics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Xinhua Hu
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Meiyu Qv
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Musaddique Hussain
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Ling-Hui Zeng
- Department of Pharmacology, Zhejiang University City College, 51 Huzhou Street, Hangzhou, 310015, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, China.
- Department of Orthopeadic Surgery of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| |
Collapse
|
50
|
Dong D, Na L, Zhou K, Wang Z, Sun Y, Zheng Q, Gao J, Zhao C, Wang W. FZD5 prevents epithelial-mesenchymal transition in gastric cancer. Cell Commun Signal 2021; 19:21. [PMID: 33618713 PMCID: PMC7898745 DOI: 10.1186/s12964-021-00708-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/18/2021] [Indexed: 12/11/2022] Open
Abstract
Background Frizzled (FZD) proteins function as receptors for WNT ligands. Members in FZD family including FZD2, FZD4, FZD7, FZD8 and FZD10 have been demonstrated to mediate cancer cell epithelial-mesenchymal transition (EMT). Methods CCLE and TCGA databases were interrogated to reveal the association of FZD5 with EMT. EMT was analyzed by investigating the alterations in CDH1 (E-cadherin), VIM (Vimentin) and ZEB1 expression, cell migration and cell morphology. Transcriptional modulation was determined by ChIP in combination with Real-time PCR. Survival was analyzed by Kaplan–Meier method. Results In contrast to other FZDs, FZD5 was identified to prevent EMT in gastric cancer. FZD5 maintains epithelial-like phenotype and is negatively modulated by transcription factors SNAI2 and TEAD1. Epithelial-specific factor ELF3 is a downstream effecter, and protein kinase C (PKC) links FZD5 to ELF3. ELF3 represses ZEB1 expression, further guarding against EMT. Moreover, FZD5 signaling requires its co-receptor LRP5 and WNT7B is a putative ligand for FZD5. FZD5 and ELF3 are associated with longer survival, whereas SNAI2 and TEAD1 are associated with shorter survival. Conclusions Taken together, FZD5-ELF3 signaling blocks EMT, and plays a potential tumor-suppressing role in gastric cancer. ![]()
Video Abstract
Collapse
Affiliation(s)
- Dan Dong
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China
| | - Lei Na
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China.,Department of Urology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Kailing Zhou
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China
| | - Zhuo Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China
| | - Yu Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China
| | - Qianqian Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China
| | - Jian Gao
- Center of Laboratory Technology and Experimental Medicine, China Medical University, Shenyang, People's Republic of China
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China.
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|