WNT4 and WNT3A Activate Cell Autonomous Wnt Signaling Independent of Secretion

Total Page:16

File Type:pdf, Size:1020Kb

WNT4 and WNT3A Activate Cell Autonomous Wnt Signaling Independent of Secretion bioRxiv preprint doi: https://doi.org/10.1101/333906; this version posted September 14, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Running Title: Secretion-independent Wnt signaling Research article WNT4 and WNT3A activate cell autonomous Wnt signaling independent of secretion Deviyani M. Rao1, Rebecca L. Ferguson1, Tomomi M. Yamamoto2, Benjamin G. Bitler2, Matthew J. Sikora1 Affiliation: 1Dept. of Pathology, 2Dept. of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus Corresponding author: Matthew J. Sikora, PhD.; Mail Stop 8104, Research Complex 1 South, Room 5117, 12801 E. 17th Ave.; Aurora, CO 80045. Tel: (303)724-4301; Fax: (303)724-3712; email: [email protected]. Twitter: @mjsikora Funding This work was supported by R00 CA193734 (MJS) and R00 CA194318 (BGB) from the National Institutes of Health, and by a grant from the Cancer League of Colorado, Inc (MJS). Authors' contributions DMR and MJS conceived of the project and experiments. DMR, RLF, and MJS designed and performed experiments. RLF, DMR, and TMY developed models for the project. DMR, RLF, BGB, and MJS contributed to data analysis and interpretation. DMR wrote the draft manuscript; all authors read and revised the manuscript, and have read and approved of this version of the manuscript. bioRxiv preprint doi: https://doi.org/10.1101/333906; this version posted September 14, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Competing interests The authors have nothing to disclose. bioRxiv preprint doi: https://doi.org/10.1101/333906; this version posted September 14, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Abstract Background: Wnt protein secretion and signaling canonically require the enzyme PORCN, suggesting PORCN inhibitors can effectively treat Wnt-dependent forms of cancer. We tested the ability of PORCN inhibitors to inhibit the growth of WNT4-dependent breast cancer cell lines, but found PORCN inhibitors did not phenocopy WNT4 knockdown. This led us to investigate the role of PORCN in WNT4 signaling. Methods: Breast cancer cell line MDA MB 134VI, fibrosarcoma cell line HT1080, and PORCN-knockout HT1080 were used as model systems for WNT4 signaling (i.e. derived from WNT4-responsive tissues; breast and bone, respectively), along with additional breast and ovarian cancer cell lines. Using these models, WNT3A or WNT4 were constitutively over-expressed. Conditioned medium was used to assess Wnt secretion and paracrine activity, and dependence on the Wnt secretion proteins PORCN and Wntless (WLS). Secreted Wnt protein activity was compared to recombinant Wnt protein. Results: Whereas WLS was universally required for Wnt secretion and paracrine signaling, the role of PORCN varied across contexts. PORCN was not required for WNT3A secretion in all cell lines, but PORCN was universally required for paracrine activity of WNT3A. In contrast, WNT4 secretion was PORCN-independent in all models tested. However, while recombinant WNT4 activated Wnt signaling in treated cells, secreted WNT4 did not present paracrine activity in any tested context. Absent the expected paracrine activity of secreted WNT4, we examined Wnt signaling directly in cells over-expressing Wnt proteins. This revealed cell autonomous activation of Wnt signaling, independent of secretion, by both WNT4 and WNT3A. Direct transfection of Wnt protein activated non-canonical Wnt signaling independent of membrane receptor activation. Conclusions: PORCN is differentially required for secretion and paracrine activity of WNT4 and WNT3A, specific to the cell type expressing Wnt proteins and the paracrine recipient of Wnt signals. Wnt proteins can activate cell autonomous signaling, independent of PORCN and of secretion. This PORCN- independent mode of Wnt signaling may be critical in cell contexts that are otherwise considered to have dysfunctional Wnt signaling. 3 bioRxiv preprint doi: https://doi.org/10.1101/333906; this version posted September 14, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Introduction Wnt signaling is an ancestrally conserved pathway that plays fundamental roles in numerous facets of embryonic development and adult tissue homeostasis. Dysregulation of Wnt signaling is a causative factor for a range of human pathologies, including several forms of cancer (reviewed in [1]). As a result, inhibition of Wnt signaling has become an attractive therapeutic target with ongoing clinical trials, with some strategies focused upstream by inhibiting the signaling actions of Wnt proteins [1–3]. Wnt proteins comprise a family of secreted glycoproteins that act as inter-cellular ligands, which stimulate a myriad of signal transduction cascades regulating cellular proliferation, stem cell renewal, cell motility, angiogenesis, and apoptosis [1, 4–6]. Wnt proteins are post-translationally modified by the O- acyltransferase Porcupine (PORCN), which palmitoylates Wnt proteins at conserved serine residues [2, 7, 8]. This lipidation forms a binding motif for interaction with Wntless (WLS), which chaperones Wnt proteins to the plasma membrane for secretion [8–10]. Once secreted, Wnt proteins signal in a paracrine manner, binding nearby receptor complexes. Wnts typically bind a Frizzled (FZD) receptor in conjunction with the LRP5 or LRP6 co-receptor, resulting in second messenger activation (Dishevelled proteins; DVL1/2/3 in humans) and initiation of either canonical (β-catenin-dependent) or non-canonical (β- catenin-independent) signaling [1, 4]. The essential initiating step in Wnt processing is palmitoylation by PORCN, which has prompted the development of PORCN inhibitors, including the IWP class of compounds [10], WNT974 (a.k.a. LGK974) [3], and others [2, 11]. These compounds have been shown to block Wnt secretion, inhibit downstream Wnt signaling, and suppress Wnt-driven tumor growth in animal models [3, 12, 13]. WNT974 is currently in Phase I/II clinical trials for cancer treatment (NCT01351103, NCT02278133). Based on these observations, PORCN inhibitors are an attractive strategy to target Wnt- driven pathologies. The Wnt protein WNT4 is critical in organogenesis of endocrine organs, regulation of bone mass, and in several steroid hormone-related phenotypes in humans [14–21]. WNT4 dysregulation via loss-of-function mutation results in developmental female to male sex reversal [22–25], and WNT4 polymorphisms are 4 bioRxiv preprint doi: https://doi.org/10.1101/333906; this version posted September 14, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. associated with endocrine dysfunction, gynecological malignancies, reduced bone density with premature skeletal aging, and related phenotypes [26–32]. WNT4 is also critical in mammary gland development, as Wnt4 knockout in mice prevents progesterone-driven ductal elongation and branching of the mammary gland during pregnancy [33, 34]. In this context, activated progesterone receptor drives expression of Wnt4 in mammary gland luminal cells resulting in paracrine signaling that supports maintenance of the mammary stem cell niche [6, 35–37]. Despite these observed critical roles of WNT4 in both normal and malignant tissues, WNT4 signaling is crudely understood due to varied context-dependent functions. Both human and murine WNT4 are reported to activate or suppress either canonical or non-canonical Wnt signaling in a context and tissue-specific manner (described in references herein). Further, conflicting reports indicate Wnt4 can or cannot activate canonical Wnt signaling in the murine mammary gland [35, 38]. It is also unclear which FZD receptor complexes are utilized by WNT4, as WNT4 is often required for distinct, non-redundant functions versus other Wnt proteins, leading to the description of WNT4 as a “problem child” among Wnts (reviewed in [34]). Based on these observations, inhibition of WNT4 signaling upstream of Wnt effector pathways, such as with PORCN inhibitors, is an attractive approach to block WNT4 signaling in a “pathway indifferent” manner and treat WNT4-related pathologies. We recently reported that regulation of WNT4 expression is co-opted by the estrogen receptor in a subtype of breast cancer, invasive lobular carcinoma (ILC) [39, 40]. Estrogen-driven WNT4 is required in ILC cells for estrogen-induced proliferation and survival, as well as anti-estrogen resistance [40]. Though WNT4-driven signaling in ILC is yet to be fully elucidated, ILC cells lack the capacity to engage canonical Wnt signaling, as the characteristic genetic loss of E-cadherin in ILC leads to loss of β-catenin protein [40, 41]. This suggests that ILC should
Recommended publications
  • Molecular and Physiological Basis for Hair Loss in Near Naked Hairless and Oak Ridge Rhino-Like Mouse Models: Tracking the Role of the Hairless Gene
    University of Tennessee, Knoxville TRACE: Tennessee Research and Creative Exchange Doctoral Dissertations Graduate School 5-2006 Molecular and Physiological Basis for Hair Loss in Near Naked Hairless and Oak Ridge Rhino-like Mouse Models: Tracking the Role of the Hairless Gene Yutao Liu University of Tennessee - Knoxville Follow this and additional works at: https://trace.tennessee.edu/utk_graddiss Part of the Life Sciences Commons Recommended Citation Liu, Yutao, "Molecular and Physiological Basis for Hair Loss in Near Naked Hairless and Oak Ridge Rhino- like Mouse Models: Tracking the Role of the Hairless Gene. " PhD diss., University of Tennessee, 2006. https://trace.tennessee.edu/utk_graddiss/1824 This Dissertation is brought to you for free and open access by the Graduate School at TRACE: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Doctoral Dissertations by an authorized administrator of TRACE: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. To the Graduate Council: I am submitting herewith a dissertation written by Yutao Liu entitled "Molecular and Physiological Basis for Hair Loss in Near Naked Hairless and Oak Ridge Rhino-like Mouse Models: Tracking the Role of the Hairless Gene." I have examined the final electronic copy of this dissertation for form and content and recommend that it be accepted in partial fulfillment of the requirements for the degree of Doctor of Philosophy, with a major in Life Sciences. Brynn H. Voy, Major Professor We have read this dissertation and recommend its acceptance: Naima Moustaid-Moussa, Yisong Wang, Rogert Hettich Accepted for the Council: Carolyn R.
    [Show full text]
  • The Extracellular Matrix Phenome Across Species
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.06.980169; this version posted March 6, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. The extracellular matrix phenome across species Cyril Statzer1 and Collin Y. Ewald1* 1 Eidgenössische Technische Hochschule Zürich, Department of Health Sciences and Technology, Institute of Translational Medicine, Schwerzenbach-Zürich CH-8603, Switzerland *Corresponding authors: [email protected] (CYE) Keywords: Phenome, genotype-to-phenotype, matrisome, extracellular matrix, collagen, data mining. Highlights • 7.6% of the human phenome originates from variations in matrisome genes • 11’671 phenotypes are linked to matrisome genes of humans, mice, zebrafish, Drosophila, and C. elegans • Expected top ECM phenotypes are developmental, morphological and structural phenotypes • Nonobvious top ECM phenotypes include immune system, stress resilience, and age-related phenotypes 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.03.06.980169; this version posted March 6, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-ND 4.0 International license. 1 Abstract 2 Extracellular matrices are essential for cellular and organismal function. Recent 3 genome-wide and phenome-wide association studies started to reveal a broad 4 spectrum of phenotypes associated with genetic variants. However, the phenome or 5 spectrum of all phenotypes associated with genetic variants in extracellular matrix 6 genes is unknown.
    [Show full text]
  • Original Article Parathyroid Hormone Induces Epithelial-To-Mesenchymal Transition Via the Wnt/Β-Catenin Signaling Pathway in Human Renal Proximal Tubular Cells
    Int J Clin Exp Pathol 2014;7(9):5978-5987 www.ijcep.com /ISSN:1936-2625/IJCEP0001621 Original Article Parathyroid hormone induces epithelial-to-mesenchymal transition via the Wnt/β-catenin signaling pathway in human renal proximal tubular cells Yunshan Guo1*, Zhen Li1*, Raohai Ding1, Hongdong Li1, Lei Zhang1, Weijie Yuan2, Yanxia Wang1 1Department of Nephrology, General Hospital of Ji’nan Military Command, Ji’nan 250031, China; 2Department of Nephrology, Shanghai Jiaotong University Affiliated First People’s Hospital, 85 Wu Jin Road, Shanghai 200080, China. *Equal contributors. Received July 30, 2014; Accepted August 21, 2014; Epub August 15, 2014; Published September 1, 2014 Abstract: Epithelial-to-mesenchymal transition (EMT) has been shown to play an important role in renal fibrogen- esis. Recent studies suggested parathyroid hormone (PTH) could accelerate EMT and subsequent organ fibrosis. However, the precise molecular mechanisms underlying PTH-induced EMT remain unknown. The present study was to investigate whether Wnt/β-catenin signaling pathway is involved in PTH-induced EMT in human renal proximal tubular cells (HK-2 cells) and to determine the profile of gene expression associated with PTH-induced EMT. PTH could induce morphological changes and gene expression characteristic of EMT in cultured HK-2 cells. Suppressing β-catenin expression or DKK1 limited gene expression characteristic of PTH-induced EMT. Based on the PCR array analysis, PTH treatment resulted in the up-regulation of 18 genes and down-regulation of 9 genes compared with the control. The results were further supported by a western blot analysis, which showed the increased Wnt4 protein expression. Wnt4 overexpression also promotes PTH-induced EMT in HK-2 cells.
    [Show full text]
  • Wnt-Independent and Wnt-Dependent Effects of APC Loss on the Chemotherapeutic Response
    International Journal of Molecular Sciences Review Wnt-Independent and Wnt-Dependent Effects of APC Loss on the Chemotherapeutic Response Casey D. Stefanski 1,2 and Jenifer R. Prosperi 1,2,3,* 1 Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46617, USA; [email protected] 2 Mike and Josie Harper Cancer Research Institute, South Bend, IN 46617, USA 3 Department of Biochemistry and Molecular Biology, Indiana University School of Medicine-South Bend, South Bend, IN 46617, USA * Correspondence: [email protected]; Tel.: +1-574-631-4002 Received: 30 September 2020; Accepted: 20 October 2020; Published: 22 October 2020 Abstract: Resistance to chemotherapy occurs through mechanisms within the epithelial tumor cells or through interactions with components of the tumor microenvironment (TME). Chemoresistance and the development of recurrent tumors are two of the leading factors of cancer-related deaths. The Adenomatous Polyposis Coli (APC) tumor suppressor is lost in many different cancers, including colorectal, breast, and prostate cancer, and its loss correlates with a decreased overall survival in cancer patients. While APC is commonly known for its role as a negative regulator of the WNT pathway, APC has numerous binding partners and functional roles. Through APC’s interactions with DNA repair proteins, DNA replication proteins, tubulin, and other components, recent evidence has shown that APC regulates the chemotherapy response in cancer cells. In this review article, we provide an overview of some of the cellular processes in which APC participates and how they impact chemoresistance through both epithelial- and TME-derived mechanisms. Keywords: adenomatous polyposis coli; chemoresistance; WNT signaling 1.
    [Show full text]
  • Tsukushi Functions As a Wnt Signaling Inhibitor by Competing with Wnt2b for Binding to Transmembrane Protein Frizzled4
    Tsukushi functions as a Wnt signaling inhibitor by competing with Wnt2b for binding to transmembrane protein Frizzled4 Kunimasa Ohtaa,b,1,2, Ayako Itoa,c,1, Sei Kuriyamaa,d,3, Giuseppe Lupoe,f, Mitsuko Kosakag,4, Shin-ichi Ohnumah, Shinichi Nakagawai, and Hideaki Tanakaa,c,d aDepartment of Developmental Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; bPrecursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan; cGlobal Center of Excellence, Kumamoto University, Kumamoto 860-8556, Japan; d21st Century Center of Excellence, Kumamoto University, Kumamoto 860-8556, Japan; eDepartment of Biology and Biotechnology “C. Darwin,” University of Rome “La Sapienza,” 00185 Rome, Italy; fIstituto Pasteur–Fondazione Cenci Bolognetti, 00185, Rome, Italy; gRIKEN Center for Developmental Biology, Kobe 650-0047, Japan; hInstitute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom; and IRIKEN Advanced Science Institute, Nakagawa RNA Biology Laboratory, Saitama 351-0198, Japan Edited* by Lynn T. Landmesser, Case Western Reserve University, Cleveland, OH, and approved July 8, 2011 (received for review January 11, 2011) The Wnt signaling pathway is essential for the development of We previously described the isolation of Tsukushi (TSK) protein diverse tissues during embryogenesis. Signal transduction is acti- isoforms (13), soluble molecules belonging to the small leucine- vated by the binding of Wnt proteins to the type I receptor low- rich proteoglycan (SLRP) family (14), and showed that they work density lipoprotein receptor–related protein 5/6 and the seven-pass as extracellular modulators of pivotal signaling cascades during transmembrane protein Frizzled (Fzd), which contains a Wnt- early embryonic development in chicks and frogs (13, 15–17).
    [Show full text]
  • The Draft Genomes of Softshell Turtle and Green Sea Turtle Yield Insights
    LETTERS OPEN The draft genomes of soft-shell turtle and green sea turtle yield insights into the development and evolution of the turtle-specific body plan Zhuo Wang1,12, Juan Pascual-Anaya2,12, Amonida Zadissa3,12, Wenqi Li4,12, Yoshihito Niimura5, Zhiyong Huang1, Chunyi Li4, Simon White3, Zhiqiang Xiong1, Dongming Fang1, Bo Wang1, Yao Ming1, Yan Chen1, Yuan Zheng1, Shigehiro Kuraku2, Miguel Pignatelli6, Javier Herrero6, Kathryn Beal6, Masafumi Nozawa7, Qiye Li1, Juan Wang1, Hongyan Zhang4, Lili Yu1, Shuji Shigenobu7, Junyi Wang1, Jiannan Liu4, Paul Flicek6, Steve Searle3, Jun Wang1,8,9, Shigeru Kuratani2, Ye Yin4, Bronwen Aken3, Guojie Zhang1,10,11 & Naoki Irie2 The unique anatomical features of turtles have raised Three major hypotheses have been proposed for the evolutionary unanswered questions about the origin of their unique body origin of turtles, including that they (i) constitute early-diverged rep- plan. We generated and analyzed draft genomes of the soft- tiles, called anapsids3, (ii) are a sister group of the lizard-snake-tuatara shell turtle (Pelodiscus sinensis) and the green sea turtle (Lepidosauria) clade4 or (iii) are closely related to a lineage that (Chelonia mydas); our results indicated the close relationship includes crocodilians and birds (Archosauria)5–8. Even using molecular of the turtles to the bird-crocodilian lineage, from which they approaches, inconsistency still remains6–9. To clarify the evolution of split ~267.9–248.3 million years ago (Upper Permian to Triassic). the turtle-specific body plan, we first addressed the question of evolu- We also found extensive expansion of olfactory receptor genes tionary origin of the turtle by performing the first genome-wide phylo- in these turtles.
    [Show full text]
  • Hippocampus Formation: an Intriguing Collaboration Henk Roelink
    bb10g06.qxd 04/03/2000 01:10 Page R279 Dispatch R279 Hippocampus formation: An intriguing collaboration Henk Roelink Recent genetic studies have shown that the signalling temporal lobes are formed. In an adult rodent, the hip- factor Wnt3a is required for formation of the pocampus is consequently still found close to the dorsal hippocampus; the developmental consequences of Wnt midline where it is initially formed (Figure 1). signalling in the hippocampus are mediated by multiple HMG-box transcription factors, with LEF-1 being It is clear that Wnt family members are required for the required just for formation of the dentate gyrus. development of many embryonic structures, functions mediated by their effects on fundamental processes such Address: Department of Biological Structure and Center for Developmental Biology, University of Washington, Box 357420, as cell proliferation, differentiation, survival or mainte- Seattle, Washington 98117-7420, USA. nance. Obtaining a clear picture of how each Wnt acts is E-mail: [email protected] complicated by the relatively large size of the family, which has at least 18 members in amniotes [3]. Analyses of Current Biology 2000, 10:R279–R281 mice lacking one or several Wnt genes have revealed some 0960-9822/00/$ – see front matter remarkable phenotypes, often characterized by failure of © 2000 Elsevier Science Ltd. All rights reserved. induction of very specific anatomical structures. Wnt3A is no exception, and although several papers have been pub- Inductive interactions are fundamental to the formation of lished already on the more obvious defects of Wnt3a all brain structures. and signalling by molecules of the knockout mice, a recent paper from McMahon and col- Wingless/Wnt family is known to play a role in many of leagues [1] has reported a very interesting defect in the them.
    [Show full text]
  • A Membrane-Associated Β-Catenin/Oct4 Complex Correlates
    DEVELOPMENT AND STEM CELLS RESEARCH ARTICLE 1171 Development 140, 1171-1183 (2013) doi:10.1242/dev.085654 © 2013. Published by The Company of Biologists Ltd A membrane-associated β-catenin/Oct4 complex correlates with ground-state pluripotency in mouse embryonic stem cells Fernando Faunes1,*, Penelope Hayward1,*, Silvia Muñoz Descalzo1, Sujash S. Chatterjee2, Tina Balayo1, Jamie Trott1, Andrew Christoforou1,3, Anna Ferrer-Vaquer4, Anna-Katerina Hadjantonakis4, Ramanuj Dasgupta2 and Alfonso Martinez Arias1,‡ SUMMARY The maintenance of pluripotency in mouse embryonic stem cells (mESCs) relies on the activity of a transcriptional network that is fuelled by the activity of three transcription factors (Nanog, Oct4 and Sox2) and balanced by the repressive activity of Tcf3. Extracellular signals modulate the activity of the network and regulate the differentiation capacity of the cells. Wnt/β-catenin signaling has emerged as a significant potentiator of pluripotency: increases in the levels of β-catenin regulate the activity of Oct4 and Nanog, and enhance pluripotency. A recent report shows that β-catenin achieves some of these effects by modulating the activity of Tcf3, and that this effect does not require its transcriptional activation domain. Here, we show that during self-renewal there is negligible transcriptional activity of β-catenin and that this is due to its tight association with membranes, where we find it in a complex with Oct4 and E-cadherin. Differentiation triggers a burst of Wnt/β-catenin transcriptional activity that coincides with the disassembly of the complex. Our results establish that β-catenin, but not its transcriptional activity, is central to pluripotency acting through a β- catenin/Oct4 complex.
    [Show full text]
  • Beta;-Catenin Signaling in Rhabdomyosarcoma
    Laboratory Investigation (2013) 93, 1090–1099 & 2013 USCAP, Inc All rights reserved 0023-6837/13 Characterization of Wnt/b-catenin signaling in rhabdomyosarcoma Srinivas R Annavarapu1,8,9, Samantha Cialfi2,9, Carlo Dominici2,3, George K Kokai4, Stefania Uccini5, Simona Ceccarelli6, Heather P McDowell2,3,7 and Timothy R Helliwell1 Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children and accounts for about 5% of all malignant paediatric tumours. b-Catenin, a multifunctional nuclear transcription factor in the canonical Wnt signaling pathway, is active in myogenesis and embryonal somite patterning. Dysregulation of Wnt signaling facilitates tumour invasion and metastasis. This study characterizes Wnt/b-catenin signaling and functional activity in paediatric embryonal and alveolar RMS. Immunohistochemical assessment of paraffin-embedded tissues from 44 RMS showed b-catenin expression in 26 cases with cytoplasmic/membranous expression in 9/14 cases of alveolar RMS, and 15/30 cases of embryonal RMS, whereas nuclear expression was only seen in 2 cases of embryonal RMS. The potential functional significance of b-catenin expression was tested in four RMS cell lines, two derived from embryonal (RD and RD18) RMS and two from alveolar (Rh4 and Rh30) RMS. Western blot analysis demonstrated the expression of Wnt-associated proteins including b-catenin, glycogen synthase kinase-3b, disheveled, axin-1, naked, LRP-6 and cadherins in all cell lines. Cell fractionation and immunofluorescence studies of the cell lines (after stimulation by human recombinant Wnt3a) showed reduced phosphorylation of b-catenin, stabilization of the active cytosolic form and nuclear translocation of b-catenin. Reporter gene assay demonstrated a T-cell factor/lymphoid-enhancing factor-mediated transactivation in these cells.
    [Show full text]
  • Wnt3a: Functions and Implications in Cancer Sha He1,2, Yi Lu1,2, Xia Liu1,2, Xin Huang1,2, Evan T
    He et al. Chin J Cancer (2015) 34:50 DOI 10.1186/s40880-015-0052-4 REVIEW Open Access Wnt3a: functions and implications in cancer Sha He1,2, Yi Lu1,2, Xia Liu1,2, Xin Huang1,2, Evan T. Keller3, Chao‑Nan Qian4* and Jian Zhang1,2,3* Abstract Wnt3a, one of Wnt family members, plays key roles in regulating pleiotropic cellular functions, including self-renewal, proliferation, differentiation, and motility. Accumulating evidence has suggested that Wnt3a promotes or suppresses tumor progression via the canonical Wnt signaling pathway depending on cancer type. In addition, the roles of Wnt3a signaling can be inhibited by multiple proteins or chemicals. Herein, we summarize the latest findings on Wnt3a as an important therapeutic target in cancer. Keywords: Wnt3a, The Wnt signaling pathway, Cancer Background: the Wnt signaling pathway cell polarity pathway (Wnt-PCP pathway) and the Wnt- The Wnt gene was first identified in 1982 and is named calcium pathway (Wnt-Ca2+ pathway) [5]. According to the Int gene in mice [1]. A subsequent study reported the characteristics of its functions, the Wnt family can that the Int gene and wingless gene in Drosophila were also be divided into two categories: Wnt1 and Wnt5a. homologous genes [2]. Therefore, both genes were rec- The Wnt1 category includes Wnt1, Wnt2, Wnt2b, Wnt3, ognized as the Wnt gene [3]. The Wnt family comprises Wnt3a, Wnt7a, Wnt8, Wnt8b, and Wnt10a, which are 19 human proteins, including Wnt1, Wnt2, Wnt2b involved in the canonical signaling pathway, whereas (Wnt13), Wnt3, Wnt3a, Wnt4, Wnt5a, Wnt5b, Wnt6, Wnt4, Wnt5a, and Wnt11 belong to the Wnt5a category Wnt7a, Wnt7b, Wnt8a, Wnt8b, Wnt9a (Wnt14), Wnt9b and activate the noncanonical signaling pathway.
    [Show full text]
  • Renal Hypodysplasia Associates with a Wnt4 Variant That Causes Aberrant Canonical Wnt Signaling
    BRIEF COMMUNICATION www.jasn.org Renal Hypodysplasia Associates with a Wnt4 Variant that Causes Aberrant Canonical Wnt Signaling † † ‡ † † Asaf Vivante,* Michal Mark-Danieli, Miriam Davidovits, Orit Harari-Steinberg, Dorit Omer, † ‡ ‡| Yehudit Gnatek, Roxana Cleper, Daniel Landau,§ Yael Kovalski, Irit Weissman,¶ †† ‡‡ || Israel Eisenstein,** Michalle Soudack, Haike Reznik Wolf, Naomi Issler,§§ Danny Lotan, †|| Yair Anikster,¶¶ and Benjamin Dekel *Department of Pediatrics, Talpiot Medical Leadership Program, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; †Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; ‡Institute of Pediatric Nephrology, Schneider Children’s Medical Center of Israel, Petah Tiqwa, Israel; §Department of Pediatrics A and Pediatric Nephrology, Saban Children’s Hospital, Soroka Medical Center, Ben Gurion University, Beer Sheva, Israel; |Children’s Medical Center, Ramla, Israel; ¶Nephrology Unit, Western Galilee Hospital, Nahariya, Israel; **Pediatric Nephrology Unit, Meyer Children’s Hospital, Rambam Health Care Campus, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel; ††Pediatric Imaging, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; ‡‡Genetic Institute, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University,
    [Show full text]
  • Wnt4/B2catenin Signaling in Medullary Kidney Myofibroblasts
    BASIC RESEARCH www.jasn.org Wnt4/b2Catenin Signaling in Medullary Kidney Myofibroblasts † †‡ | Derek P. DiRocco,* Akio Kobayashi,* Makoto M. Taketo,§ Andrew P. McMahon, and †‡ Benjamin D. Humphreys* *Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts; †Harvard Medical School, Boston, Massachusetts; ‡Harvard Stem Cell Institute, Cambridge, Massachusetts; §Department of Pharmacology, Graduate School of Medicine, Kyoto University, Yoshida-Konoé-cho, Sakyo, Kyoto, Japan; and |Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California ABSTRACT Injury to the adult kidney induces a number of developmental genes thought to regulate repair, including Wnt4. During kidney development, early nephron precursors and medullary stroma both express Wnt4, where it regulates epithelialization and controls smooth muscle fate, respectively. Expression patterns and roles for Wnt4 in the adult kidney, however, remain unclear. In this study, we used reporters, lineage analysis, and conditional knockout or activation of the Wnt/b-catenin pathway to investigate Wnt4 in the adult kidney. Proliferating, medullary, interstitial myofibroblasts strongly expressed Wnt4 during renal fibrosis, whereas tubule epithelia, except for the collecting duct, did not. Exogenous Wnt4 drove myofi- broblast differentiation of a pericyte-like cell line, suggesting that Wnt4 might regulate pericyte-to-myo- fibroblast transition through autocrine signaling. However, conditional deletion of Wnt4 in interstitial cells did not reduce myofibroblast proliferation, cell number, or myofibroblast gene expression during fibrosis. Because the injured kidney expresses multiple Wnt ligands that might compensate for the absence of Wnt4, we generated a mouse model with constitutive activation of canonical Wnt/b-catenin signaling in interstitial pericytes and fibroblasts.
    [Show full text]