7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      To condense or not to condense: Wnt regulation by centrosome-nucleated biomolecular condensates

      article-commentary

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Colorectal cancer is the third leading cause of cancer deaths worldwide, and, thus, understanding genetic changes driving cancer development and progression is critical. A key breakthrough occurred 31 y ago when scientists cloned the tumor suppressor gene adenomatous polyposis coli (APC), mutated in families with a predisposition to colon cancer. Subsequent work revealed that APC is part of the destruction complex (DC), a multiprotein machine that negatively regulates a key developmental signaling pathway, the Wnt pathway (1). Wnt signaling regulates cell fate choice and adult stem cell maintenance in all animals. However, key questions remain about the mechanisms by which the DC works. In PNAS, Lach et al. (2) provide exciting insights. Because of the power of Wnt signaling, evolution crafted mechanisms to keep signaling firmly off in the ligand’s absence. Combined work in Drosophila, mice, and cultured colorectal cancer cells provided important insights (3). The key event is regulating stability of the Wnt effector β-catenin (βcat; Fig. 1A ). When present, it works with TCF family transcription factors to activate Wnt target genes. To keep the pathway off, a multiprotein complex including APC, another large scaffolding protein—Axin—and two protein kinases—glycogen synthase kinase-3 (GSK3) and casein kinase 1-alpha (CK1)—captures and phosphorylates βcat, and then delivers it to an E3 ubiquitin ligase, targeting it for proteolytic destruction. In tissues where signaling is active, Wnt ligands bind a multiprotein receptor that turns down DC activity, thus stabilizing cytoplasmic and nuclear βcat and activating gene expression. In APC mutant colorectal cancers, βcat is stabilized even when signaling is off, activating signaling constitutively. Fig. 1. Centrosomes nucleate formation of Wnt regulatory DCs. (A) Canonical Wnt regulation. (B) When Axin is overexpressed in cells, it forms multiple puncta. (C) Hypothetical multiprotein DC biomolecular condensate. (D) Axin expressed at endogenous levels assembles into one or two puncta colocalizing with centrosomes. A–C were produced by laboratory alumna Kristi Schaefer; D is from Max Wilson. This textbook description sounds tidy, leaving the impression that the Wnt regulatory mechanism is solved. However, key questions remain. For example, Axin alone can act as a scaffold to bring the two kinases to βcat (4), leaving APC’s function unclear. More puzzling, a series of studies suggested that a four-protein DC is not the whole story. When Axin is expressed in cultured cells, it assembles into multiple “puncta,” into which are recruited APC, the kinases, and βcat (e.g., refs. 5 and 6; Fig. 1B ). Axin’s DIX domain mediates protein polymerization (7), potentially driving assembly of puncta containing many copies of each DC protein (Fig. 1C ). This suggested that the DC might be a biomolecular condensate (reviewed in ref. 8). These large, non-membrane-bound structures assemble by multivalent protein interactions, some mediated by intrinsically disordered regions, thus generating large protein complexes by “liquid–liquid phase separation” (9). DC proteins can phase separate in vitro (e.g., ref. 10). Further, elevated resolution microscopy revealed that Axin puncta seen in cultured mammalian cells when Axin levels are elevated are structured multiprotein complexes (11, 12). Similar complexes are seen in vivo in Drosophila embryos, with Axin expressed only a few-fold above endogenous levels, and their subcellular localization and assembly are regulated by upstream Wnt signals (13), suggesting that they are functional. More than 100 Axins assemble in each punctum. However, other groups took alternate approaches, building DCs from the bottom up from purified proteins, and defining molecular interactions involved in assembly (14, 15). This was quite successful, reconstituting, in one case, not only βcat phosphorylation but also ubiquitination by the E3 ligase. The in vitro protein complexes were substantially smaller than those seen in vivo, with ≤10 Axins in each. These data raised a key question: What is the true assembly state of the DC in vivo? Lach et al. (2) take this on via a highly multidisciplinary approach. They note that phase-separating systems can exhibit switch-like responses to changes in concentration, potentially explaining differences in DC assembly in vitro and in vivo. This occurs, in part, because newly synthesized Axin or APC proteins must choose between joining an existing complex or nucleating a new one. Their concentrations and posttranslational modifications determine their location in a high-dimensional phase diagram, governing whether these proteins remain diffuse, spontaneously demix into existing droplets, or nucleate new condensates. Thus, precisely controlling protein expression levels is key to defining the true assembly state. To visualize DCs at endogenous levels, Lach et al. tagged the endogenous βcat gene with a fluorescent protein, and visualized cells live. βcat accumulated at the plasma membrane in cadherin-based cell adhesion complexes, while cytoplasmic levels were low in the absence of Wnt signaling. Wnt signaling stabilized cytoplasmic βcat, as expected. What was less expected was that, in the absence of Wnt signals, βcat also accumulated in one or two bright, spherical puncta near the nuclei (2). These resembled liquid-like biomolecular condensates, fusing and splitting. Importantly, puncta stability was regulated by Wnt signaling: Adding Wnt ligands led to loss of puncta. To confirm this, Lach et al. used an optogenetically controlled Wnt receptor, which was activated by light. Activating the receptor also led to puncta dissolution. Similar puncta were observed with Axin tagged at its endogenous locus, eliminating worries about overexpression (Fig. 1D ). The assembled puncta contain all DC proteins, which could come in and out of puncta readily and rapidly, consistent with known biomolecular condensate properties. Together, these data suggested the puncta are the active DC. Most intriguing, the one or two puncta per cell colocalized with centrosomes, the cell’s major microtubule organizing center (2). Lach et al. confirmed that puncta and centrosomes exclusively colocalized when Axin was expressed at endogenous levels, but, when they used inducible expression to elevate Axin levels, multiple extracentrosomal DC puncta formed. These data suggested centrosomes regulate DC assembly by serving as nucleation sites. At the lower endogenous levels of Axin, centrosomes might locally concentrate Axin or other DC proteins enough to trigger Axin multimerization and thus phase transition. Lach et al. first tested this by mathematically modeling DC assembly and function, contrasting predicted rates of βcat phosphorylation with or without a “nucleator.” Strikingly, their models predicted that nucleation strongly accelerates βcat phosphorylation over a wide range of parameters, improving DC efficiency. Lach et al. (2) then created a tool to test whether condensation elevates Wnt regulatory activity in vivo: a version of the DC kinase GSK3 that oligomerizes in response to light. Strikingly, inducing GSK3 oligomerization rapidly increased GSK3 concentration specifically in centrosomal puncta, without triggering assembly of extracentrosomal puncta. Further, GSK3 oligomerization prevented Wnt signaling from turning down the DC, and inhibited Wnt-induced human embryonic stem cell differentiation, putting the work into the context of normal Wnt signaling. Together, this work provides key insights into Wnt signaling, helping reconcile earlier in vitro, cell culture and in vivo experiments concerning the DC assembly state (2). These data reveal that, at least in cultured mammalian cells, DCs form phase-separated biomolecular condensates even when proteins are all expressed at endogenous levels. Lach et al.'s modeling suggests this increases efficiency of targeting βcat for phosphorylation and thus destruction. This work also suggests that earlier experiments suggesting the presence of many such puncta likely resulted from Axin overexpression, confirming the very tight concentration threshold for condensate formation. In their system, centrosomes nucleate condensates, triggering condensation just below the normal concentration threshold. This aspect will have broader relevance, as similar concentration-dependent behaviors and nucleation mechanisms may regulate other important biomolecular condensates. This work (2) also raises many exciting questions. Which protein interactions mediate centrosomal recruitment? Which molecular interactions then trigger condensation? Candidates include Axin polymerization, APC’s Armadillo repeats, and APC’s enigmatic CID/R2-B motif (11, 16, 17). We also must define how APC protein truncations seen in colorectal tumors affect condensate assembly or function. The data open questions about mechanisms that regulate condensate. It was striking that GSK3 oligomerization rendered DCs immune to disassembly by Wnt signaling—in addition to phosphorylating βcat, GSK3 also phosphorylates Axin and APC, and GSK3 phosphorylation of APC’s CID/R2-B motif may regulate assembly (11). Handoff to the E3 ubiquitin ligase is the next step in targeting βcat for destruction. We must determine whether and how the E3 ligase is recruited to centrosomal condensates. The E3 F-box protein TrCP can localize to centrosomes (18), and in vitro work supports physical interaction of E3 ligase and DC proteins (12, 19). While localization of individual DC proteins to centrosomes was known, the centrosome’s role in nucleation was a surprise. However, it opens new questions. Both mouse embryos (20) and Drosophila (21) lacking centrioles can survive without noticeable defects in Wnt signaling, revealing that centrosomes are not essential for this process. What other cellular structures might serve as nucleation sites? Finally, as the authors note (2), it will be important to use their system in partnership with in vitro approaches to define how Wnt signaling acts through the Wnt receptor complex to activate Dishevelled and turn down DC activity. It will be exciting to see how complementary approaches help tackle these key issues in cell, developmental, and cancer biology.

          Related collections

          Most cited references21

          • Record: found
          • Abstract: found
          • Article: not found

          Biomolecular condensates: organizers of cellular biochemistry

          In addition to membrane-bound organelles, eukaryotic cells feature various membraneless compartments, including the centrosome, the nucleolus and various granules. Many of these compartments form through liquid–liquid phase separation, and the principles, mechanisms and regulation of their assembly as well as their cellular functions are now beginning to emerge.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities.

            The WNT signal transduction cascade is a main regulator of development throughout the animal kingdom. Wnts are also key drivers of most types of tissue stem cells in adult mammals. Unsurprisingly, mutated Wnt pathway components are causative to multiple growth-related pathologies and to cancer. Here, we describe the core Wnt/β-catenin signaling pathway, how it controls stem cells, and contributes to disease. Finally, we discuss strategies for Wnt-based therapies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Flies without centrioles.

              Centrioles and centrosomes have an important role in animal cell organization, but it is uncertain to what extent they are essential for animal development. The Drosophila protein DSas-4 is related to the human microcephaly protein CenpJ and the C. elegans centriolar protein Sas-4. We show that DSas-4 is essential for centriole replication in flies. DSas-4 mutants start to lose centrioles during embryonic development, and, by third-instar larval stages, no centrioles or centrosomes are detectable. Mitotic spindle assembly is slow in mutant cells, and approximately 30% of the asymmetric divisions of larval neuroblasts are abnormal. Nevertheless, mutant flies develop with near normal timing into morphologically normal adults. These flies, however, have no cilia or flagella and die shortly after birth because their sensory neurons lack cilia. Thus, centrioles are essential for the formation of centrosomes, cilia, and flagella, but, remarkably, they are not essential for most aspects of Drosophila development.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc Natl Acad Sci U S A
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                30 September 2022
                11 October 2022
                30 March 2023
                : 119
                : 41
                : e2213905119
                Affiliations
                [1] aDepartment of Biology, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599-3280;
                [2] bLineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599
                Author notes

                Author contributions: M.P. wrote the paper.

                Article
                202213905
                10.1073/pnas.2213905119
                9565305
                36179040
                50c5ce04-07b2-40bf-95b8-78621dfd1c5b
                Copyright © 2022 the Author(s). Published by PNAS.

                This article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 3
                Funding
                Funded by: HHS | NIH | National Institute of General Medical Sciences (NIGMS) 100000057
                Award ID: R35 GM118096
                Award Recipient : Mark Peifer
                Categories
                408
                437
                Commentary
                Biological Sciences
                Biophysics and Computational Biology

                Comments

                Comment on this article