1
|
Kamizaki K, Katsukawa M, Yamamoto A, Fukada SI, Uezumi A, Endo M, Minami Y. Ror2 signaling regulated by differential Wnt proteins determines pathological fate of muscle mesenchymal progenitors. Cell Death Dis 2024; 15:784. [PMID: 39468010 PMCID: PMC11519583 DOI: 10.1038/s41419-024-07173-9] [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: 07/02/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Skeletal muscle mesenchymal progenitors (MPs) play a critical role in supporting muscle regeneration. However, under pathological conditions, they contribute to intramuscular adipose tissue accumulation, involved in muscle diseases, including muscular dystrophy and sarcopenia, age-related muscular atrophy. How MP fate is determined in these different contexts remains unelucidated. Here, we report that Ror2, a non-canonical Wnt signaling receptor, is selectively expressed in MPs and regulates their pathological features in a differential ligand-dependent manner. We identified Wnt11 and Wnt5b as ligands of Ror2. In vitro, Wnt11 inhibited MP senescence, which is required for normal muscle regeneration, and Wnt5b promoted MP proliferation. We further found that both Wnts are abundant in degenerating muscle and synergistically stimulate Ror2, leading to unwanted MP proliferation and eventually intramuscular adipose tissue accumulation. These findings provide evidence that Ror2-mediated signaling elicited by differential Wnts plays a critical role in determining the pathological fate of MPs.
Collapse
Affiliation(s)
- Koki Kamizaki
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Mitsuko Katsukawa
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Ayano Yamamoto
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - So-Ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akiyoshi Uezumi
- Division of Cell Heterogeneity, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Mitsuharu Endo
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan.
| |
Collapse
|
2
|
Shi DL. Canonical and Non-Canonical Wnt Signaling Generates Molecular and Cellular Asymmetries to Establish Embryonic Axes. J Dev Biol 2024; 12:20. [PMID: 39189260 PMCID: PMC11348223 DOI: 10.3390/jdb12030020] [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: 06/11/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
The formation of embryonic axes is a critical step during animal development, which contributes to establishing the basic body plan in each particular organism. Wnt signaling pathways play pivotal roles in this fundamental process. Canonical Wnt signaling that is dependent on β-catenin regulates the patterning of dorsoventral, anteroposterior, and left-right axes. Non-canonical Wnt signaling that is independent of β-catenin modulates cytoskeletal organization to coordinate cell polarity changes and asymmetric cell movements. It is now well documented that components of these Wnt pathways biochemically and functionally interact to mediate cell-cell communications and instruct cellular polarization in breaking the embryonic symmetry. The dysfunction of Wnt signaling disrupts embryonic axis specification and proper tissue morphogenesis, and mutations of Wnt pathway genes are associated with birth defects in humans. This review discusses the regulatory roles of Wnt pathway components in embryonic axis formation by focusing on vertebrate models. It highlights current progress in decoding conserved mechanisms underlying the establishment of asymmetry along the three primary body axes. By providing an in-depth analysis of canonical and non-canonical pathways in regulating cell fates and cellular behaviors, this work offers insights into the intricate processes that contribute to setting up the basic body plan in vertebrate embryos.
Collapse
Affiliation(s)
- De-Li Shi
- Department of Medical Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China;
- Laboratory of Developmental Biology, Centre National de la Recherche Scientifique (CNRS), UMR7622, Institut de Biologie Paris-Seine (IBPS), Sorbonne University, 75005 Paris, France
| |
Collapse
|
3
|
Kamizaki K, Minami Y, Nishita M. Role of the Ror family receptors in Wnt5a signaling. In Vitro Cell Dev Biol Anim 2024; 60:489-501. [PMID: 38587578 DOI: 10.1007/s11626-024-00885-4] [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: 10/30/2023] [Accepted: 01/30/2024] [Indexed: 04/09/2024]
Abstract
Ror-family receptors, Ror1 and Ror2, are type I transmembrane proteins that possess an extracellular cysteine-rich domain, which is conserved throughout the Frizzled-family receptors and is a binding site for Wnt ligands. Both Ror1 and Ror2 function primarily as receptors or co-receptors for Wnt5a to activate the β-catenin-independent, non-canonical Wnt signaling, thereby regulating cell polarity, migration, proliferation, and differentiation depending on the context. Ror1 and Ror2 are expressed highly in many tissues during embryogenesis but minimally or scarcely in adult tissues, with some exceptions. In contrast, Ror1 and Ror2 are expressed in many types of cancers, and their high expression often contributes to the progression of the disease. Therefore, Ror1 and Ror2 have been proposed as potential targets for the treatment of the malignancies. In this review, we provide an overview of the regulatory mechanisms of Ror1/Ror2 expression and discuss how Wnt5a-Ror1/Ror2 signaling is mediated and regulated by their interacting proteins.
Collapse
Affiliation(s)
- Koki Kamizaki
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Michiru Nishita
- Department of Biochemistry, Fukushima Medical University School of Medicine, 1 Hikariga-Oka, Fukushima, 960-1295, Japan.
| |
Collapse
|
4
|
Ito T. Molecular pathology of small cell lung cancer: Overview from studies on neuroendocrine differentiation regulated by ASCL1 and Notch signaling. Pathol Int 2024; 74:239-251. [PMID: 38607250 DOI: 10.1111/pin.13426] [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: 10/18/2023] [Revised: 03/20/2024] [Accepted: 03/24/2024] [Indexed: 04/13/2024]
Abstract
Pulmonary neuroendocrine (NE) cells are rare airway epithelial cells. The balance between Achaete-scute complex homolog 1 (ASCL1) and hairy and enhancer of split 1, one of the target molecules of the Notch signaling pathway, is crucial for NE differentiation. Small cell lung cancer (SCLC) is a highly aggressive lung tumor, characterized by rapid cell proliferation, a high metastatic potential, and the acquisition of resistance to treatment. The subtypes of SCLC are defined by the expression status of NE cell-lineage transcription factors, such as ASCL1, which roles are supported by SRY-box 2, insulinoma-associated protein 1, NK2 homeobox 1, and wingless-related integration site signaling. This network reinforces NE differentiation and may induce the characteristic morphology and chemosensitivity of SCLC. Notch signaling mediates cell-fate decisions, resulting in an NE to non-NE fate switch. The suppression of NE differentiation may change the histological type of SCLC to a non-SCLC morphology. In SCLC with NE differentiation, Notch signaling is typically inactive and genetically or epigenetically regulated. However, Notch signaling may be activated after chemotherapy, and, in concert with Yes-associated protein signaling and RE1-silencing transcription factor, suppresses NE differentiation, producing intratumor heterogeneity and chemoresistance. Accumulated information on the molecular mechanisms of SCLC will contribute to further advances in the control of this recalcitrant cancer.
Collapse
Grants
- 20H03691 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
- 18K19489 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
- 16590318 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
- 25460439 Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology of Japan
- Smoking Research Foundation, Japan
Collapse
Affiliation(s)
- Takaaki Ito
- Department of Medical Technology, Kumamoto Health Science University Faculty of Health Sciences, Kumamoto, Japan
- Department of Pathology and Experimental Medicine, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
5
|
Yu EPY, Saxena V, Perin S, Ekker M. Loss of dlx5a/ dlx6a Locus Alters Non-Canonical Wnt Signaling and Meckel's Cartilage Morphology. Biomolecules 2023; 13:1347. [PMID: 37759750 PMCID: PMC10526740 DOI: 10.3390/biom13091347] [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: 08/05/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
The dlx genes encode transcription factors that establish a proximal-distal polarity within neural crest cells to bestow a regional identity during craniofacial development. The expression regions of dlx paralogs are overlapping yet distinct within the zebrafish pharyngeal arches and may also be involved in progressive morphologic changes and organization of chondrocytes of the face. However, how each dlx paralog of dlx1a, dlx2a, dlx5a and dlx6a affects craniofacial development is still largely unknown. We report here that the average lengths of the Meckel's, palatoquadrate and ceratohyal cartilages in different dlx mutants were altered. Mutants for dlx5a-/- and dlx5i6-/-, where the entire dlx5a/dlx6a locus was deleted, have the shortest lengths for all three structures at 5 days post fertilization (dpf). This phenotype was also observed in 14 dpf larvae. Loss of dlx5i6 also resulted in increased proliferation of neural crest cells and expression of chondrogenic markers. Additionally, altered expression and function of non-canonical Wnt signaling were observed in these mutants suggesting a novel interaction between dlx5i6 locus and non-canonical Wnt pathway regulating ventral cartilage morphogenesis.
Collapse
Affiliation(s)
| | | | | | - Marc Ekker
- Department of Biology, University of Ottawa, Marie-Curie Private, Ottawa, ON K1N 94A, Canada (S.P.)
| |
Collapse
|
6
|
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
|
7
|
Dranow DB, Le Pabic P, Schilling TF. The non-canonical Wnt receptor Ror2 is required for cartilage cell polarity and morphogenesis of the craniofacial skeleton in zebrafish. Development 2023; 150:dev201273. [PMID: 37039156 PMCID: PMC10163346 DOI: 10.1242/dev.201273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/21/2023] [Indexed: 04/12/2023]
Abstract
Non-canonical/β-catenin-independent Wnt signaling plays crucial roles in tissue/cell polarity in epithelia, but its functions have been less well studied in mesenchymal tissues, such as the skeleton. Mutations in non-canonical Wnt signaling pathway genes cause human skeletal diseases such as Robinow syndrome and Brachydactyly Type B1, which disrupt bone growth throughout the endochondral skeleton. Ror2 is one of several non-canonical Wnt receptor/co-receptors. Here, we show that ror2-/- mutant zebrafish have craniofacial skeletal defects, including disruptions of chondrocyte polarity. ror1-/- mutants appear to be phenotypically wild type, but loss of both ror1 and ror2 leads to more severe cartilage defects, indicating partial redundancy. Skeletal defects in ror1/2 double mutants resemble those of wnt5b-/- mutants, suggesting that Wnt5b is the primary Ror ligand in zebrafish. Surprisingly, the proline-rich domain of Ror2, but not its kinase domain, is required to rescue its function in mosaic transgenic experiments in ror2-/- mutants. These results suggest that endochondral bone defects in ROR-related human syndromes reflect defects in cartilage polarity and morphogenesis.
Collapse
Affiliation(s)
- Daniel B. Dranow
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Pierre Le Pabic
- Department of Biology & Marine Biology, University of North Carolina, Wilmington, NC 28403, USA
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
8
|
Caetano da Silva C, Ostertag A, Raman R, Muller M, Cohen-Solal M, Collet C. wnt11f2 Zebrafish, an Animal Model for Development and New Insights in Bone Formation. Zebrafish 2023; 20:1-9. [PMID: 36795617 PMCID: PMC9968865 DOI: 10.1089/zeb.2022.0042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Wnt signaling is a key regulator of osteoblast differentiation and mineralization in humans and animals, mediated by the canonical Wnt/β-catenin and noncanonical signaling pathways. Both pathways are crucial in regulating osteoblastogenesis and bone formation. The zebrafish silberblick (slb) carries a mutation in wnt11f2, a gene that contributes to embryonic morphogenesis; however, its role in bone morphology is unknown. wnt11f2 was originally known as wnt11; it was recently reclassified to avoid confusion in comparative genetics and disease modeling. The goal of this review is to summarize the characterization of the wnt11f2 zebrafish mutant and to deliver some new insights concerning its role in skeletal development. In addition to the previously described defects in early development in this mutant as well as craniofacial dysmorphia, we show an increase in tissue mineral density in the heterozygous mutant that points to a possible role of wnt11f2 in high bone mass phenotypes.
Collapse
Affiliation(s)
- Caroline Caetano da Silva
- INSERM U1132 and Université Paris-Cité, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris, France
| | - Agnes Ostertag
- INSERM U1132 and Université Paris-Cité, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris, France
| | - Ratish Raman
- Laboratory for Organogenesis and Regeneration (LOR), GIGA-Research, Liège University, Liège, Belgium
| | - Marc Muller
- Laboratory for Organogenesis and Regeneration (LOR), GIGA-Research, Liège University, Liège, Belgium
| | - Martine Cohen-Solal
- INSERM U1132 and Université Paris-Cité, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris, France
| | - Corinne Collet
- INSERM U1132 and Université Paris-Cité, Reference Centre for Rare Bone Diseases, Hospital Lariboisière, Paris, France.,UF de Génétique Moléculaire, Hôpital Robert Debré, APHP, Paris, France
| |
Collapse
|
9
|
Yoon J, Sun J, Lee M, Hwang YS, Daar IO. Wnt4 and ephrinB2 instruct apical constriction via Dishevelled and non-canonical signaling. Nat Commun 2023; 14:337. [PMID: 36670115 PMCID: PMC9860048 DOI: 10.1038/s41467-023-35991-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 01/11/2023] [Indexed: 01/22/2023] Open
Abstract
Apical constriction is a cell shape change critical to vertebrate neural tube closure, and the contractile force required for this process is generated by actin-myosin networks. The signaling cue that instructs this process has remained elusive. Here, we identify Wnt4 and the transmembrane ephrinB2 protein as playing an instructive role in neural tube closure as members of a signaling complex we termed WERDS (Wnt4, EphrinB2, Ror2, Dishevelled (Dsh2), and Shroom3). Disruption of function or interaction among members of the WERDS complex results in defects of apical constriction and neural tube closure. The mechanism of action involves an interaction of ephrinB2 with the Dsh2 scaffold protein that enhances the formation of the WERDS complex, which in turn, activates Rho-associated kinase to induce apical constriction. Moreover, the ephrinB2/Dsh2 interaction promotes non-canonical Wnt signaling and shows how cross-talk between two major signal transduction pathways, Eph/ephrin and Wnt, coordinate morphogenesis of the neural tube.
Collapse
Affiliation(s)
- Jaeho Yoon
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| | - Jian Sun
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Moonsup Lee
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Yoo-Seok Hwang
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Ira O Daar
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| |
Collapse
|
10
|
Shi DL. Wnt/planar cell polarity signaling controls morphogenetic movements of gastrulation and neural tube closure. Cell Mol Life Sci 2022; 79:586. [PMID: 36369349 PMCID: PMC11803072 DOI: 10.1007/s00018-022-04620-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/25/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Abstract
Gastrulation and neurulation are successive morphogenetic processes that play key roles in shaping the basic embryonic body plan. Importantly, they operate through common cellular and molecular mechanisms to set up the three spatially organized germ layers and to close the neural tube. During gastrulation and neurulation, convergent extension movements driven by cell intercalation and oriented cell division generate major forces to narrow the germ layers along the mediolateral axis and elongate the embryo in the anteroposterior direction. Apical constriction also makes an important contribution to promote the formation of the blastopore and the bending of the neural plate. Planar cell polarity proteins are major regulators of asymmetric cell behaviors and critically involved in a wide variety of developmental processes, from gastrulation and neurulation to organogenesis. Mutations of planar cell polarity genes can lead to general defects in the morphogenesis of different organs and the co-existence of distinct congenital diseases, such as spina bifida, hearing deficits, kidney diseases, and limb elongation defects. This review outlines our current understanding of non-canonical Wnt signaling, commonly known as Wnt/planar cell polarity signaling, in regulating morphogenetic movements of gastrulation and neural tube closure during development and disease. It also attempts to identify unanswered questions that deserve further investigations.
Collapse
Affiliation(s)
- De-Li Shi
- Institute of Medical Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Laboratory of Developmental Biology, CNRS-UMR7622, Institut de Biologie Paris-Seine (IBPS), Sorbonne University, Paris, France.
| |
Collapse
|
11
|
De Jong HN, Dewey FE, Cordero P, Victorio RA, Kirillova A, Huang Y, Madhvani R, Seo K, Werdich AA, Lan F, Orcholski M, Liu WR, Erbilgin A, Wheeler MT, Chen R, Pan S, Kim YM, Bommakanti K, Marcou CA, Bos JM, Haddad F, Ackerman M, Vasan RS, MacRae C, Wu JC, de Jesus Perez V, Snyder M, Parikh VN, Ashley EA. Wnt Signaling Interactor WTIP (Wilms Tumor Interacting Protein) Underlies Novel Mechanism for Cardiac Hypertrophy. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2022; 15:e003563. [PMID: 35671065 PMCID: PMC10445530 DOI: 10.1161/circgen.121.003563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/15/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The study of hypertrophic cardiomyopathy (HCM) can yield insight into the mechanisms underlying the complex trait of cardiac hypertrophy. To date, most genetic variants associated with HCM have been found in sarcomeric genes. Here, we describe a novel HCM-associated variant in the noncanonical Wnt signaling interactor WTIP (Wilms tumor interacting protein) and provide evidence of a role for WTIP in complex disease. METHODS In a family affected by HCM, we used exome sequencing and identity-by-descent analysis to identify a novel variant in WTIP (p.Y233F). We knocked down WTIP in isolated neonatal rat ventricular myocytes with lentivirally delivered short hairpin ribonucleic acids and in Danio rerio via morpholino injection. We performed weighted gene coexpression network analysis for WTIP in human cardiac tissue, as well as association analysis for WTIP variation and left ventricular hypertrophy. Finally, we generated induced pluripotent stem cell-derived cardiomyocytes from patient tissue, characterized size and calcium cycling, and determined the effect of verapamil treatment on calcium dynamics. RESULTS WTIP knockdown caused hypertrophy in neonatal rat ventricular myocytes and increased cardiac hypertrophy, peak calcium, and resting calcium in D rerio. Network analysis of human cardiac tissue indicated WTIP as a central coordinator of prohypertrophic networks, while common variation at the WTIP locus was associated with human left ventricular hypertrophy. Patient-derived WTIP p.Y233F-induced pluripotent stem cell-derived cardiomyocytes recapitulated cellular hypertrophy and increased resting calcium, which was ameliorated by verapamil. CONCLUSIONS We demonstrate that a novel genetic variant found in a family with HCM disrupts binding to a known Wnt signaling protein, misregulating cardiomyocyte calcium dynamics. Further, in orthogonal model systems, we show that expression of the gene WTIP is important in complex cardiac hypertrophy phenotypes. These findings, derived from the observation of a rare Mendelian disease variant, uncover a novel disease mechanism with implications across diverse forms of cardiac hypertrophy.
Collapse
Affiliation(s)
| | | | - Pablo Cordero
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Rachelle A. Victorio
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Anna Kirillova
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Yong Huang
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Roshni Madhvani
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Kinya Seo
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Andreas A. Werdich
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Feng Lan
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Mark Orcholski
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - W. Robert Liu
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Ayca Erbilgin
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Matthew T. Wheeler
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Rui Chen
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Stephen Pan
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Young M. Kim
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Krishna Bommakanti
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Cherisse A. Marcou
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - J. Martijn Bos
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Francois Haddad
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Michael Ackerman
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Ramachandran S. Vasan
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Calum MacRae
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Joseph C. Wu
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Vinicio de Jesus Perez
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | - Michael Snyder
- Department of Genetics (H.N.D., R.C., M.S., E.A.A.), Department of Medicine (F.E.D., A.K., Y.H., R.M., K.S., F.L., M.O., W.R.L., A.E., M.T.W., S.P., Y.M.K., K.B., F.H., J.C.W., V.d.J.P., V.N.P., E.A.A.), and Biomedical Informatics (P.C.), Stanford University, CA. Brigham and Women’s Hospital, Harvard University, Boston, MA (R.A.V., A.A.W., C.M.). Mayo Clinic, Rochester, MN (C.A.M., J.M.B., M.A.). Boston University School of Medicine, MA (R.S.V.)
| | | | | |
Collapse
|
12
|
Endo M, Kamizaki K, Minami Y. The Ror-Family Receptors in Development, Tissue Regeneration and Age-Related Disease. Front Cell Dev Biol 2022; 10:891763. [PMID: 35493090 PMCID: PMC9043558 DOI: 10.3389/fcell.2022.891763] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/29/2022] [Indexed: 12/17/2022] Open
Abstract
The Ror-family proteins, Ror1 and Ror2, act as receptors or co-receptors for Wnt5a and its related Wnt proteins to activate non-canonical Wnt signaling. Ror1 and/or Ror2-mediated signaling plays essential roles in regulating cell polarity, migration, proliferation and differentiation during developmental morphogenesis, tissue-/organo-genesis and regeneration of adult tissues following injury. Ror1 and Ror2 are expressed abundantly in developing tissues in an overlapping, yet distinct manner, and their expression in adult tissues is restricted to specific cell types such as tissue stem/progenitor cells. Expression levels of Ror1 and/or Ror2 in the adult tissues are increased following injury, thereby promoting regeneration or repair of these injured tissues. On the other hand, disruption of Wnt5a-Ror2 signaling is implicated in senescence of tissue stem/progenitor cells that is related to the impaired regeneration capacity of aged tissues. In fact, Ror1 and Ror2 are implicated in age-related diseases, including tissue fibrosis, atherosclerosis (or arteriosclerosis), neurodegenerative diseases, and cancers. In these diseases, enhanced and/or sustained (chronic) expression of Ror1 and/or Ror2 is observed, and they might contribute to the progression of these diseases through Wnt5a-dependent and -independent manners. In this article, we overview recent advances in our understanding of the roles of Ror1 and Ror2-mediated signaling in the development, tissue regeneration and age-related diseases, and discuss their potential to be therapeutic targets for chronic inflammatory diseases and cancers.
Collapse
|
13
|
Saraswathy VM, Kurup AJ, Sharma P, Polès S, Poulain M, Fürthauer M. The E3 Ubiquitin Ligase Mindbomb1 controls planar cell polarity-dependent convergent extension movements during zebrafish gastrulation. eLife 2022; 11:71928. [PMID: 35142609 PMCID: PMC8937233 DOI: 10.7554/elife.71928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Vertebrate Delta/Notch signaling involves multiple ligands, receptors and transcription factors. Delta endocytosis - a critical event for Notch activation - is however essentially controlled by the E3 Ubiquitin ligase Mindbomb1 (Mib1). Mib1 inactivation is therefore often used to inhibit Notch signaling. However, recent findings indicate that Mib1 function extends beyond the Notch pathway. We report a novel Notch-independent role of Mib1 in zebrafish gastrulation. mib1 null mutants and morphants display impaired Convergence Extension (CE) movements. Comparison of different mib1 mutants and functional rescue experiments indicate that Mib1 controls CE independently of Notch. Mib1-dependent CE defects can be rescued using the Planar Cell Polarity (PCP) downstream mediator RhoA, or enhanced through knock-down of the PCP ligand Wnt5b. Mib1 regulates CE through its RING Finger domains that have been implicated in substrate ubiquitination, suggesting that Mib1 may control PCP protein trafficking. Accordingly, we show that Mib1 controls the endocytosis of the PCP component Ryk and that Ryk internalization is required for CE. Numerous morphogenetic processes involve both Notch and PCP signaling. Our observation that during zebrafish gastrulation Mib1 exerts a Notch-independent control of PCP-dependent CE movements suggest that Mib1 loss of function phenotypes should be cautiously interpreted depending on the biological context.
Collapse
Affiliation(s)
| | | | | | - Sophie Polès
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | | | | |
Collapse
|
14
|
Menck K, Heinrichs S, Wlochowitz D, Sitte M, Noeding H, Janshoff A, Treiber H, Ruhwedel T, Schatlo B, von der Brelie C, Wiemann S, Pukrop T, Beißbarth T, Binder C, Bleckmann A. WNT11/ROR2 signaling is associated with tumor invasion and poor survival in breast cancer. J Exp Clin Cancer Res 2021; 40:395. [PMID: 34911552 PMCID: PMC8672621 DOI: 10.1186/s13046-021-02187-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer has been associated with activation of the WNT signaling pathway, although no driver mutations in WNT genes have been found yet. Instead, a high expression of the alternative WNT receptor ROR2 was observed, in particular in breast cancer brain metastases. However, its respective ligand and downstream signaling in this context remained unknown. METHODS We modulated the expression of ROR2 in human breast cancer cells and characterized their gene and protein expression by RNA-Seq, qRT-PCR, immunoblots and reverse phase protein array (RPPA) combined with network analyses to understand the molecular basis of ROR2 signaling in breast cancer. Using co-immunoprecipitations, we verified the interaction of ROR2 with the identified ligand, WNT11. The functional consequences of WNT11/ROR2 signaling for tumor cell aggressiveness were assessed by microscopy, impedance sensing as well as viability and invasion assays. To evaluate the translational significance of our findings, we performed gene set enrichment, expression and survival analyses on human breast cancer brain metastases. RESULTS We found ROR2 to be highly expressed in aggressive breast tumors and associated with worse metastasis-free survival. ROR2 overexpression induced a BRCAness-like phenotype in a cell-context specific manner and rendered cells resistant to PARP inhibition. High levels of ROR2 were furthermore associated with defects in cell morphology and cell-cell-contacts leading to increased tumor invasiveness. On a molecular level, ROR2 overexpression upregulated several non-canonical WNT ligands, in particular WNT11. Co-immunoprecipitation confirmed that WNT11 indeed interacts with the cysteine-rich domain of ROR2 and triggers its invasion-promoting signaling via RHO/ROCK. Knockdown of WNT11 reversed the pro-invasive phenotype and the cellular changes in ROR2-overexpressing cells. CONCLUSIONS Taken together, our study revealed a novel auto-stimulatory loop in which ROR2 triggers the expression of its own ligand, WNT11, resulting in enhanced tumor invasion associated with breast cancer metastasis.
Collapse
Affiliation(s)
- Kerstin Menck
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149, Münster, Germany
- West German Cancer Center, University Hospital Münster, 48149, Münster, Germany
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Saskia Heinrichs
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149, Münster, Germany
- West German Cancer Center, University Hospital Münster, 48149, Münster, Germany
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Darius Wlochowitz
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Maren Sitte
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Helen Noeding
- Institute for Physical Chemistry, Georg August University Göttingen, 37075, Göttingen, Germany
| | - Andreas Janshoff
- Institute for Physical Chemistry, Georg August University Göttingen, 37075, Göttingen, Germany
| | - Hannes Treiber
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075, Göttingen, Germany
| | - Bawarjan Schatlo
- Department of Neurosurgery, University Medical Center Göttingen, 37099, Göttingen, Germany
| | | | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Tobias Pukrop
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Claudia Binder
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149, Münster, Germany.
- West German Cancer Center, University Hospital Münster, 48149, Münster, Germany.
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany.
| |
Collapse
|
15
|
Creighton JH, Jessen JR. Core pathway proteins and the molecular basis of planar polarity in the zebrafish gastrula. Semin Cell Dev Biol 2021; 125:17-25. [PMID: 34635444 DOI: 10.1016/j.semcdb.2021.09.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 11/30/2022]
Abstract
The planar polarization of cells and subcellular structures is critical for embryonic development. Coordination of this polarity can provide cells a sense of direction in relation to the anterior-posterior and dorsal-ventral body axes. Fly epithelia use a core pathway comprised of transmembrane (Van Gogh/Strabismus, Frizzled, and Flamingo/Starry night) and cytoplasmic (Prickle or Spiny-legs, Dishevelled, and Diego) proteins to communicate directional information between cells and thereby promote the uniform orientation of structures such as hairs. In the zebrafish gastrula, planar polarity underlies complex cellular processes, including directed migration and intercalation, that are required to shape the embryo body. Like other vertebrates, the zebrafish genome encodes homologs of each core protein, and it is well-established that polarized gastrula cell behaviors are regulated by some of them. However, it is unknown whether a conserved six-member core protein pathway regulates planar polarity during zebrafish gastrulation. Here, we review our current understanding of core protein function as it relates to two specific examples of planar polarity, the dorsal convergence of lateral gastrula cells and the mediolateral intercalation of midline cells. We consider the hallmarks of fly planar polarity and discuss data regarding asymmetric protein localization and function, and the intercellular communication of polarity information.
Collapse
Affiliation(s)
- Joy H Creighton
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, USA
| | - Jason R Jessen
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, USA.
| |
Collapse
|
16
|
Ka C, Gautam S, Marshall SR, Tice LP, Martinez-Bartolome M, Fenner JL, Range RC. Receptor Tyrosine Kinases ror1/2 and ryk Are Co-expressed with Multiple Wnt Signaling Components During Early Development of Sea Urchin Embryos. THE BIOLOGICAL BULLETIN 2021; 241:140-157. [PMID: 34706206 PMCID: PMC11257382 DOI: 10.1086/715237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
AbstractA combination of receptors, co-receptors, and secreted Wnt modulators form protein complexes at the cell surface that activate one or more of the three different Wnt signaling pathways (Wnt/β-catenin, Wnt/JNK, and Wnt/Ca2+). Two or more of these pathways are often active in the same cellular territories, forming Wnt signaling networks; however, the molecular mechanisms necessary to integrate information from these pathways in these situations are unclear in any in vivo model system. Recent studies have implicated two Wnt binding receptor tyrosine kinases, receptor tyrosine kinase-like orphan receptor (Ror) and related-to-receptor tyrosine kinase (Ryk), in the regulation of canonical and non-canonical Wnt signaling pathways, depending on the context; however, the spatiotemporal expression of these genes in relation to Wnt signaling components has not been well characterized in most deuterostome model systems. Here we use a combination of phylogenetic and spatiotemporal gene expression analyses to characterize Ror and Ryk orthologs in sea urchin embryos. Our phylogenetic analysis indicates that both ror1/2 and ryk originated as single genes from the metazoan ancestor. Expression analyses indicate that ror1/2 and ryk are expressed in the same domains of many Wnt ligands and Frizzled receptors essential for the specification and patterning of germ layers along the early anterior-posterior axis. In addition, both genes are co-expressed with Wnt signaling components in the gut, ventral ectoderm, and anterior neuroectoderm territories later in development. Together, our results indicate that Ror and Ryk have a complex evolutionary history and that their spatiotemporal expression suggests that they could contribute to the complexity of Wnt signaling in early sea urchin embryogenesis.
Collapse
Affiliation(s)
- C Ka
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - S Gautam
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - SR Marshall
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS 39762
| | - LP Tice
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS 39762
| | | | - JL Fenner
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| | - RC Range
- Department of Biological Sciences, Auburn University, Auburn, Alabama 36849
| |
Collapse
|
17
|
Dawes ML, Soeller C, Scholpp S. Studying molecular interactions in the intact organism: fluorescence correlation spectroscopy in the living zebrafish embryo. Histochem Cell Biol 2020; 154:507-519. [PMID: 33067656 PMCID: PMC7609432 DOI: 10.1007/s00418-020-01930-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
Abstract
Cell behaviour and function is determined through the interactions of a multitude of molecules working in concert. To observe these molecular dynamics, biophysical studies have been developed that track single interactions. Fluorescence correlation spectroscopy (FCS) is an optical biophysical technique that non-invasively resolves single molecules through recording the signal intensity at the femtolitre scale. However, recording the behaviour of these biomolecules using in vitro-based assays often fails to recapitulate the full range of variables in vivo that directly confer dynamics. Therefore, there has been an increasing interest in observing the state of these biomolecules within living organisms such as the zebrafish Danio rerio. In this review, we explore the advancements of FCS within the zebrafish and compare and contrast these findings to those found in vitro.
Collapse
Affiliation(s)
- Michael L Dawes
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX4 4QD, UK
| | - Christian Soeller
- Living Systems Institute, College of Engineering, Mathematics, and Physical Sciences, University of Exeter, Exeter, EX4 4QD, UK
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX4 4QD, UK.
| |
Collapse
|
18
|
Zhang H, Rong X, Wang C, Liu Y, Lu L, Li Y, Zhao C, Zhou J. VBP1 modulates Wnt/β-catenin signaling by mediating the stability of the transcription factors TCF/LEFs. J Biol Chem 2020; 295:16826-16839. [PMID: 32989053 PMCID: PMC7864075 DOI: 10.1074/jbc.ra120.015282] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/23/2020] [Indexed: 12/29/2022] Open
Abstract
The Wnt/β-catenin pathway is one of the major pathways that regulates embryonic development, adult homeostasis, and stem cell self-renewal. In this pathway, transcription factors T-cell factor and lymphoid enhancer factor (TCF/LEF) serve as a key switch to repress or activate Wnt target gene transcription by recruiting repressor molecules or interacting with the β-catenin effector, respectively. It has become evident that the protein stability of the TCF/LEF family members may play a critical role in controlling the activity of the Wnt/β-catenin signaling pathway. However, factors that regulate the stability of TCF/LEFs remain largely unknown. Here, we report that pVHL binding protein 1 (VBP1) regulates the Wnt/β-catenin signaling pathway by controlling the stability of TCF/LEFs. Surprisingly, we found that either overexpression or knockdown of VBP1 decreased Wnt/β-catenin signaling activity in both cultured cells and zebrafish embryos. Mechanistically, VBP1 directly binds to all four TCF/LEF family members and von Hippel-Lindau tumor-suppressor protein (pVHL). Either overexpression or knockdown of VBP1 increases the association between TCF/LEFs and pVHL and then decreases the protein levels of TCF/LEFs via proteasomal degradation. Together, our results provide mechanistic insights into the roles of VBP1 in controlling TCF/LEFs protein stability and regulating Wnt/β-catenin signaling pathway activity.
Collapse
Affiliation(s)
- Haifeng Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiaozhi Rong
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Caixia Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Ling Lu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yun Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Chengtian Zhao
- Institute of Evolution and Marine Biodiversity and College of Marine Biology, Ocean University of China, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jianfeng Zhou
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
19
|
Abstract
Gastrulation is a critical early morphogenetic process of animal development, during which the three germ layers; mesoderm, endoderm and ectoderm, are rearranged by internalization movements. Concurrent epiboly movements spread and thin the germ layers while convergence and extension movements shape them into an anteroposteriorly elongated body with head, trunk, tail and organ rudiments. In zebrafish, gastrulation follows the proliferative and inductive events that establish the embryonic and extraembryonic tissues and the embryonic axis. Specification of these tissues and embryonic axes are controlled by the maternal gene products deposited in the egg. These early maternally controlled processes need to generate sufficient cell numbers and establish the embryonic polarity to ensure normal gastrulation. Subsequently, after activation of the zygotic genome, the zygotic gene products govern mesoderm and endoderm induction and germ layer patterning. Gastrulation is initiated during the maternal-to-zygotic transition, a process that entails both activation of the zygotic genome and downregulation of the maternal transcripts. Genomic studies indicate that gastrulation is largely controlled by the zygotic genome. Nonetheless, genetic studies that investigate the relative contributions of maternal and zygotic gene function by comparing zygotic, maternal and maternal zygotic mutant phenotypes, reveal significant contribution of maternal gene products, transcripts and/or proteins, that persist through gastrulation, to the control of gastrulation movements. Therefore, in zebrafish, the maternally expressed gene products not only set the stage for, but they also actively participate in gastrulation morphogenesis.
Collapse
Affiliation(s)
- Lilianna Solnica-Krezel
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
20
|
Roberts JL, Liu G, Paglia DN, Kinter CW, Fernandes LM, Lorenzo J, Hansen MF, Arif A, Drissi H. Deletion of
Wnt5a
in osteoclasts results in bone loss through decreased bone formation. Ann N Y Acad Sci 2020; 1463:45-59. [DOI: 10.1111/nyas.14293] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/25/2019] [Accepted: 12/04/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Joseph L. Roberts
- Department of Orthopaedics Emory University School of Medicine Atlanta Georgia
| | - Guanglu Liu
- Department of Orthopaedics Emory University School of Medicine Atlanta Georgia
| | - David N. Paglia
- Department of Orthopaedics, New Jersey Medical School Rutgers University Newark New Jersey
| | | | | | - Joseph Lorenzo
- Department of Medicine and Department of Orthopaedic Surgery University of Connecticut Health Farmington Connecticut
| | - Marc F. Hansen
- Center for Molecular Medicine University of Connecticut Health Farmington Connecticut
| | - Abul Arif
- Department of Orthopaedics Emory University School of Medicine Atlanta Georgia
| | - Hicham Drissi
- Department of Orthopaedics Emory University School of Medicine Atlanta Georgia
| |
Collapse
|
21
|
Williams ML, Solnica-Krezel L. Cellular and molecular mechanisms of convergence and extension in zebrafish. Curr Top Dev Biol 2020; 136:377-407. [DOI: 10.1016/bs.ctdb.2019.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
22
|
Tenjin Y, Kudoh S, Kubota S, Yamada T, Matsuo A, Sato Y, Ichimura T, Kohrogi H, Sashida G, Sakagami T, Ito T. Ascl1-induced Wnt11 regulates neuroendocrine differentiation, cell proliferation, and E-cadherin expression in small-cell lung cancer and Wnt11 regulates small-cell lung cancer biology. J Transl Med 2019; 99:1622-1635. [PMID: 31231131 DOI: 10.1038/s41374-019-0277-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/03/2019] [Accepted: 05/08/2019] [Indexed: 01/09/2023] Open
Abstract
The involvement of Wnt signaling in human lung cancer remains unclear. This study investigated the role of Wnt11 in neuroendocrine (NE) differentiation, cell proliferation, and epithelial-to-mesenchymal transition (EMT) in human small-cell lung cancer (SCLC). Immunohistochemical staining of resected specimens showed that Wnt11 was expressed at higher levels in SCLCs than in non-SCLCs; 58.8% of SCLC, 5.2% of adenocarcinoma (ADC), and 23.5% of squamous cell carcinoma tissues stained positive for Wnt11. A positive relationship was observed between Achaete-scute complex homolog 1 (Ascl1) and Wnt11 expression in SCLC cell lines, and this was supported by transcriptome data from SCLC tissue. The expression of Wnt11 and some NE markers increased after the transfection of ASCL1 into the A549 ADC cell line. Knockdown of Ascl1 downregulated Wnt11 expression in SCLC cell lines. Ascl1 regulated Wnt11 expression via lysine H3K27 acetylation at the enhancer region of the WNT11 gene. Wnt11 controlled NE differentiation, cell proliferation, and E-cadherin expression under the regulation of Ascl1 in SCLC cell lines. The phosphorylation of AKT and p38 mitogen-activated protein kinase markedly increased after transfection of WNT11 into the SBC3 SCLC cell line, which suggests that Wnt11 promotes cell proliferation in SCLC cell lines. Ascl1 plays an important role in regulating the Wnt signaling pathway and is one of the driver molecules of Wnt11 in human SCLC. Ascl1 and Wnt11 may employ a cooperative mechanism to control the biology of SCLC. The present results indicate the therapeutic potential of targeting the Ascl1-Wnt11 signaling axis and support the clinical utility of Wnt11 as a biological marker in SCLC.
Collapse
Affiliation(s)
- Yuki Tenjin
- Department of Pathology and Experimental Medicine, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan.,Department of Respiratory Medicine, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Shinji Kudoh
- Department of Pathology and Experimental Medicine, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Sho Kubota
- Laboratory of Transcriptional Regulation in Leukemogenesis, International Research Center for Medical Sciences, Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Tatsuya Yamada
- Department of Pathology and Experimental Medicine, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan.,Department of Thoracic Surgery, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Akira Matsuo
- Department of Pathology and Experimental Medicine, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Younosuke Sato
- Department of Pathology and Experimental Medicine, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takaya Ichimura
- Department of Pathology, Faculty of Medicine, Saitama Medical University, Saitama, 350-0495, Japan
| | - Hirotsugu Kohrogi
- Department of Respiratory Medicine, Omuta Tenryo Hospital, Tenryo 1-100, Omuta, Fukuoka, 836-8556, Japan
| | - Goro Sashida
- Laboratory of Transcriptional Regulation in Leukemogenesis, International Research Center for Medical Sciences, Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Takaaki Ito
- Department of Pathology and Experimental Medicine, Graduate School of Medical Science, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto, 860-8556, Japan.
| |
Collapse
|
23
|
Li Y, Han X, Xu W, Rao Z, Li X. Purification and characterization of the extracellular region of human receptor tyrosine kinase like orphan receptor 2 (ROR2). Protein Expr Purif 2019; 158:74-80. [PMID: 30826310 DOI: 10.1016/j.pep.2019.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 11/25/2022]
Abstract
Receptor tyrosine kinase like orphan receptor 2 (ROR2) is a co-receptor for some Wnt proteins including Wnt5a that activate the noncanonical Wnt/planar cell polarity (PCP) signaling pathway. Upregulation of ROR2 is associated with several cancer forms. The extracellular region of ROR2, which contains an immunoglobulin (Ig)-like domain, a Frizzled like cysteine-rich domain (CRD) and a Kringle domain, is a potential anticancer drug target. The structural and biochemical properties of the ROR2 extracellular region remain largely unexplored. Here we describe the mapping and purification, using a baculovirus - insect cell system, of a near-full-length ROR2 extracellular fragment (residues 53-402), which is well-behaved and suitable for future structural and biochemical analysis. We show that the extracellular region of ROR2 per se is monomeric in solution. Different monoclonal antibodies raised against the purified ROR2 protein can specifically recognize the protein and can either inhibit or activate the PCP activity in a cell-based assay, and are thus potentially useful for future mechanistic and therapeutic/diagnostic studies. The biological relevance of these antibodies further demonstrates that the purified recombinant ROR2 protein is properly folded and biochemically active.
Collapse
Affiliation(s)
- Yuan Li
- Collage of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xu Han
- Collage of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Wenqing Xu
- Department of Biological Structure, University of Washington, Seattle, WA, 98195, USA.
| | - Zihe Rao
- Collage of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Xin Li
- Collage of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
24
|
Bai Y, Liu C, Zhou J, Rong X, Wang H. Molecular, functional, and gene expression analysis of zebrafish Ror1 receptor. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:355-363. [PMID: 30242697 DOI: 10.1007/s10695-018-0567-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 08/29/2018] [Indexed: 06/08/2023]
Abstract
Ror family of receptor tyrosine kinases ROR1 and ROR2 plays crucial roles in animal development by regulating cell proliferation, differentiation, and migration, as well as survival and death by acting as a receptor or co-receptor for Wnt5a and mediating Wnt5a-induced activation. Compared with our extensive understanding of ROR2, our knowledge of ROR1 is limited. In this study, we characterized the zebrafish ror1 gene and determined its temporal and spatial expression and biological activity. Sequence comparison and phylogenetic analyses indicate that its protein structure is similar to its mammalian orthologs. During embryogenesis, the ror1 mRNA levels were relatively low or undetectable at 6 and 9 h postfertilization. In adult fish, ror1 mRNA was most abundantly expressed in the ovary and testis. The levels of ror1 mRNA in non-reproductive system tissues were very low or barely detectable. Spatiotemporal distribution of ror1 and its ligand wnt5a in the ovary was then investigated. Reverse transcription PCR on isolated follicle layers and denuded oocytes demonstrated that both wnt5a and ror1 were exclusively expressed in the oocyte but not in the follicle layers. During oogenesis, the ror1 mRNA levels were relatively low from I to IV stage oocytes and increased dramatically at V stage oocyte. Unlike ror1, the wnt5a mRNA levels were increased gradually from I to V stage oocyte. When Ror1 was co-transfected with Wnt5a and Wnt3a in HEK293T cells, the Wnt3a-induced Wnt reporter activity was inhibited by Ror1 in a dose-dependent manner. Taken together, these results provide new information about the structural and functional conservation, spatial and temporal expression, and biological activity of Ror1 in a fish model organism.
Collapse
Affiliation(s)
- Yan Bai
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Chengdong Liu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Jianfeng Zhou
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Xiaozhi Rong
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
| | - Hongying Wang
- Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, Key Laboratory of State Ethnic Affairs Commission for Biological Technology, College of Life Sciences, South-Central University for Nationalities, Wuhan, 430074, China.
| |
Collapse
|
25
|
Ripp C, Loth J, Petrova I, Linnemannstöns K, Ulepic M, Fradkin L, Noordermeer J, Wodarz A. Drosophila Ror is a nervous system-specific co-receptor for Wnt ligands. Biol Open 2018; 7:bio.033001. [PMID: 30341100 PMCID: PMC6262871 DOI: 10.1242/bio.033001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Wnt ligands are secreted glycoproteins that control many developmental processes and are crucial for homeostasis of numerous tissues in the adult organism. Signal transduction of Wnts involves the binding of Wnts to receptor complexes at the surface of target cells. These receptor complexes are commonly formed between a member of the Frizzled family of seven-pass transmembrane proteins and a co-receptor, which is usually a single-pass transmembrane protein. Among these co-receptors are several with structural homology to receptor tyrosine kinases, including Ror, PTK7, Ryk and MUSK. In vertebrates, Ror-2 and PTK7 are important regulators of planar cell polarity (PCP). By contrast, PCP phenotypes were not reported for mutations in off-track (otk) and off-track2 (otk2), encoding the Drosophila orthologs of PTK7. Here we show that Drosophila Ror is expressed in the nervous system and localizes to the plasma membrane of perikarya and neurites. A null allele of Ror is homozygous viable and fertile, does not display PCP phenotypes and interacts genetically with mutations in otk and otk2. We show that Ror binds specifically to Wingless (Wg), Wnt4 and Wnt5 and also to Frizzled2 (Fz2) and Otk. Our findings establish Drosophila Ror as a Wnt co-receptor expressed in the nervous system. Summary:Drosophila Ror is a Wnt co-receptor expressed in the nervous system. A Ror null mutant allele is viable and shows genetic interaction with mutations in off-track and off-track2.
Collapse
Affiliation(s)
- Caroline Ripp
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Julia Loth
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Iveta Petrova
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Karen Linnemannstöns
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Monique Ulepic
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Kerpener Str. 62, 50937 Köln, Germany
| | - Lee Fradkin
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2300RC Leiden, The Netherlands.,Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, LRB 760, Worcester, MA 01605, USA
| | - Jasprien Noordermeer
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Andreas Wodarz
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany .,Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Kerpener Str. 62, 50937 Köln, Germany.,Cluster of Excellence - Cellular stress response in aging-associated diseases (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| |
Collapse
|
26
|
Hosseini V, Dani C, Geranmayeh MH, Mohammadzadeh F, Nazari Soltan Ahmad S, Darabi M. Wnt lipidation: Roles in trafficking, modulation, and function. J Cell Physiol 2018; 234:8040-8054. [PMID: 30341908 DOI: 10.1002/jcp.27570] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/17/2018] [Indexed: 12/30/2022]
Abstract
The Wnt signaling pathway consists of various downstream target proteins that have substantial roles in mammalian cell proliferation, differentiation, and development. Its aberrant activity can lead to uncontrolled proliferation and tumorigenesis. The posttranslational connection of fatty acyl chains to Wnt proteins provides the unique capacity for regulation of Wnt activity. In spite of the past belief that Wnt molecules are subject to dual acylation, it has been shown that these proteins have only one acylation site and undergo monounsaturated fatty acylation. The Wnt monounsaturated fatty acyl chain is more than just a hydrophobic coating and appears to be critical for Wnt signaling, transport, and receptor activation. Here, we provide an overview of recent findings in Wnt monounsaturated fatty acylation and the mechanism by which this lipid moiety regulates Wnt activity from the site of production to its receptor interactions.
Collapse
Affiliation(s)
- Vahid Hosseini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hossein Geranmayeh
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Mohammadzadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Masoud Darabi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| |
Collapse
|
27
|
Azimian-Zavareh V, Hossein G, Ebrahimi M, Dehghani-Ghobadi Z. Wnt11 alters integrin and cadherin expression by ovarian cancer spheroids and inhibits tumorigenesis and metastasis. Exp Cell Res 2018; 369:90-104. [PMID: 29753625 DOI: 10.1016/j.yexcr.2018.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 04/20/2018] [Accepted: 05/08/2018] [Indexed: 12/14/2022]
Abstract
The present study investigated the role of Wnt11 in multicellular tumor spheroid-like structures (MCTS) ovarian cancer cell proliferation, migration and invasion in vitro and in vivo tumorigenesis and metastasis in xenograft nude mice model. Moreover, samples from human serous ovarian cancer (SOC) were used to assess the association of Wnt11 with integrins and cadherins. The data showed that Wnt11 overexpressing SKOV-3 cells became more compact accompanied by increased expression of E-and N-cadherin and lower expression of EpCAM and CD44. The α5, β2, β3 and β6 integrin subunits expression levels were significantly reduced in Wnt11 overexpressing cells accompanied with significantly reduced disaggregation of Wnt11 overexpressing SKOV-3 MCTS on ECM components. Moreover, Wnt11 overexpressing SKOV-3 MCTS showed decreased migration, invasion as well as no tumor growth and metastasis in vivo. We found that Wnt11 significantly and negatively correlated with ITGB2, ITGB6, and EpCAM and positively with CDH-1 in high-grade SOC specimens. Our results suggest that Wnt11 impedes MCTS attachment to ECM components and therefore can affect ovarian cancer progression.
Collapse
Affiliation(s)
- Vajihe Azimian-Zavareh
- Department of Animal Physiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Ghamartaj Hossein
- Department of Animal Physiology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Zeinab Dehghani-Ghobadi
- Department of Animal Physiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
28
|
Mattes B, Dang Y, Greicius G, Kaufmann LT, Prunsche B, Rosenbauer J, Stegmaier J, Mikut R, Özbek S, Nienhaus GU, Schug A, Virshup DM, Scholpp S. Wnt/PCP controls spreading of Wnt/β-catenin signals by cytonemes in vertebrates. eLife 2018; 7:36953. [PMID: 30060804 PMCID: PMC6086664 DOI: 10.7554/elife.36953] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/16/2018] [Indexed: 12/31/2022] Open
Abstract
Signaling filopodia, termed cytonemes, are dynamic actin-based membrane structures that regulate the exchange of signaling molecules and their receptors within tissues. However, how cytoneme formation is regulated remains unclear. Here, we show that Wnt/planar cell polarity (PCP) autocrine signaling controls the emergence of cytonemes, and that cytonemes subsequently control paracrine Wnt/β-catenin signal activation. Upon binding of the Wnt family member Wnt8a, the receptor tyrosine kinase Ror2 becomes activated. Ror2/PCP signaling leads to the induction of cytonemes, which mediate the transport of Wnt8a to neighboring cells. In the Wnt-receiving cells, Wnt8a on cytonemes triggers Wnt/β-catenin-dependent gene transcription and proliferation. We show that cytoneme-based Wnt transport operates in diverse processes, including zebrafish development, murine intestinal crypt and human cancer organoids, demonstrating that Wnt transport by cytonemes and its control via the Ror2 pathway is highly conserved in vertebrates. Communication helps the cells that make up tissues and organs to work together as a team. One way that cells share information with each other as tissues grow and develop is by exchanging signaling proteins. These interact with receptors on the surface of other cells; this causes the cell to change how it behaves. The Wnt family of signaling proteins orchestrate organ development. Wnt proteins influence which types of cells develop, how fast they divide, and how and when they move. Relatively few cells, or small groups of cells, in developing tissues produce Wnt proteins, while larger groups nearby respond to the signals. We do not fully understand how Wnt proteins travel between cells, but recent work revealed an unexpected mechanism – cells seem to hand-deliver their messages. Finger-like structures called cytonemes grow out of the cell membrane and carry Wnt proteins to their destination. If the cytonemes do not form properly the target cells do not behave correctly, which can lead to severe tissue malformation. Mattes et al. have now investigated how cytonemes form using a combination of state-of-the-art genetic and high-resolution imaging techniques. In initial experiments involving zebrafish cells that were grown in the laboratory, Mattes et al. found that the Wnt proteins kick start their own transport; before they travel to their destination, they act on the cells that made them. A Wnt protein called Wnt8a activates the receptor Ror2 on the surface of the signal-producing cell. Ror2 then triggers signals inside the cell that begin the assembly of the cytonemes. The more Ror2 is activated, the more cytonemes the cell makes, and the more Wnt signals it can send out. This mechanism operates in various tissues: Ror2 also controls the cytoneme transport process in living zebrafish embryos, the mouse intestine and human stomach tumors. This knowledge will help researchers to develop new ways to control Wnt signaling, which could help to produce new treatments for diseases ranging from cancers (for example in the stomach and bowel) to degenerative diseases such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Benjamin Mattes
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Yonglong Dang
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Gediminas Greicius
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Benedikt Prunsche
- Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Jakob Rosenbauer
- John von Neumann Institute for Computing, Jülich Supercomputing Centre, Jülich, Germany
| | - Johannes Stegmaier
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Imaging and Computer Vision, RWTH Aachen University, Aachen, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Suat Özbek
- Centre of Organismal Studies, University of Heidelberg, Karlsruhe, Germany
| | - Gerd Ulrich Nienhaus
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Applied Physics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Nanotechnology, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Department of Physics, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Alexander Schug
- John von Neumann Institute for Computing, Jülich Supercomputing Centre, Jülich, Germany.,Steinbuch Centre for Computing, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - David M Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
29
|
Frizzled-8 integrates Wnt-11 and transforming growth factor-β signaling in prostate cancer. Nat Commun 2018; 9:1747. [PMID: 29717114 PMCID: PMC5931552 DOI: 10.1038/s41467-018-04042-w] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 03/29/2018] [Indexed: 01/14/2023] Open
Abstract
Wnt-11 promotes cancer cell migration and invasion independently of β-catenin but the receptors involved remain unknown. Here, we provide evidence that FZD8 is a major Wnt-11 receptor in prostate cancer that integrates Wnt-11 and TGF-β signals to promote EMT. FZD8 mRNA is upregulated in multiple prostate cancer datasets and in metastatic cancer cell lines in vitro and in vivo. Analysis of patient samples reveals increased levels of FZD8 in cancer, correlating with Wnt-11. FZD8 co-localizes and co-immunoprecipitates with Wnt-11 and potentiates Wnt-11 activation of ATF2-dependent transcription. FZD8 silencing reduces prostate cancer cell migration, invasion, three-dimensional (3D) organotypic cell growth, expression of EMT-related genes, and TGF-β/Smad-dependent signaling. Mechanistically, FZD8 forms a TGF-β-regulated complex with TGF-β receptors that is mediated by the extracellular domains of FZD8 and TGFBR1. Targeting FZD8 may therefore inhibit aberrant activation of both Wnt and TGF-β signals in prostate cancer. Wnt11 has been shown to play a role in invasion and metastasis of prostate cancer. Here the authors show that in prostate cancer cells Wnt11 signals through the Fzd8 receptor and report an interaction between Fzd8 and TGF-β receptors regulating the transcription of a subset of TGF-beta genes.
Collapse
|
30
|
Susman MW, Karuna EP, Kunz RC, Gujral TS, Cantú AV, Choi SS, Jong BY, Okada K, Scales MK, Hum J, Hu LS, Kirschner MW, Nishinakamura R, Yamada S, Laird DJ, Jao LE, Gygi SP, Greenberg ME, Ho HYH. Kinesin superfamily protein Kif26b links Wnt5a-Ror signaling to the control of cell and tissue behaviors in vertebrates. eLife 2017; 6:e26509. [PMID: 28885975 PMCID: PMC5590807 DOI: 10.7554/elife.26509] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022] Open
Abstract
Wnt5a-Ror signaling constitutes a developmental pathway crucial for embryonic tissue morphogenesis, reproduction and adult tissue regeneration, yet the molecular mechanisms by which the Wnt5a-Ror pathway mediates these processes are largely unknown. Using a proteomic screen, we identify the kinesin superfamily protein Kif26b as a downstream target of the Wnt5a-Ror pathway. Wnt5a-Ror, through a process independent of the canonical Wnt/β-catenin-dependent pathway, regulates the cellular stability of Kif26b by inducing its degradation via the ubiquitin-proteasome system. Through this mechanism, Kif26b modulates the migratory behavior of cultured mesenchymal cells in a Wnt5a-dependent manner. Genetic perturbation of Kif26b function in vivo caused embryonic axis malformations and depletion of primordial germ cells in the developing gonad, two phenotypes characteristic of disrupted Wnt5a-Ror signaling. These findings indicate that Kif26b links Wnt5a-Ror signaling to the control of morphogenetic cell and tissue behaviors in vertebrates and reveal a new role for regulated proteolysis in noncanonical Wnt5a-Ror signal transduction.
Collapse
Affiliation(s)
- Michael W Susman
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| | - Edith P Karuna
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Ryan C Kunz
- Department of Cell BiologyHarvard Medical SchoolBostonUnited States
| | - Taranjit S Gujral
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
- Division of Human BiologyFred Hutchinson Cancer Research CenterSeattleUnited States
| | - Andrea V Cantú
- Department of Obstetrics, Gynecology and Reproductive SciencesCenter for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of CaliforniaSan FranciscoUnited States
| | - Shannon S Choi
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Brigette Y Jong
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Kyoko Okada
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Michael K Scales
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Jennie Hum
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Linda S Hu
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| | - Marc W Kirschner
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
| | - Ryuichi Nishinakamura
- Department of Kidney DevelopmentInstitute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Soichiro Yamada
- Department of Biomedical EngineeringUniversity of CaliforniaDavisUnited States
| | - Diana J Laird
- Department of Obstetrics, Gynecology and Reproductive SciencesCenter for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of CaliforniaSan FranciscoUnited States
| | - Li-En Jao
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| | - Steven P Gygi
- Department of Cell BiologyHarvard Medical SchoolBostonUnited States
| | | | - Hsin-Yi Henry Ho
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
- Department of Cell Biology and Human AnatomyUniversity of California, Davis School of MedicineDavisUnited States
| |
Collapse
|
31
|
Bayerlová M, Menck K, Klemm F, Wolff A, Pukrop T, Binder C, Beißbarth T, Bleckmann A. Ror2 Signaling and Its Relevance in Breast Cancer Progression. Front Oncol 2017; 7:135. [PMID: 28695110 PMCID: PMC5483589 DOI: 10.3389/fonc.2017.00135] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/07/2017] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is a heterogeneous disease and has been classified into five molecular subtypes based on gene expression profiles. Signaling processes linked to different breast cancer molecular subtypes and different clinical outcomes are still poorly understood. Aberrant regulation of Wnt signaling has been implicated in breast cancer progression. In particular Ror1/2 receptors and several other members of the non-canonical Wnt signaling pathway were associated with aggressive breast cancer behavior. However, Wnt signals are mediated via multiple complex pathways, and it is clinically important to determine which particular Wnt cascades, including their domains and targets, are deregulated in poor prognosis breast cancer. To investigate activation and outcome of the Ror2-dependent non-canonical Wnt signaling pathway, we overexpressed the Ror2 receptor in MCF-7 and MDA-MB231 breast cancer cells, stimulated the cells with its ligand Wnt5a, and we knocked-down Ror1 in MDA-MB231 cells. We measured the invasive capacity of perturbed cells to assess phenotypic changes, and mRNA was profiled to quantify gene expression changes. Differentially expressed genes were integrated into a literature-based non-canonical Wnt signaling network. The results were further used in the analysis of an independent dataset of breast cancer patients with metastasis-free survival annotation. Overexpression of the Ror2 receptor, stimulation with Wnt5a, as well as the combination of both perturbations enhanced invasiveness of MCF-7 cells. The expression-responsive targets of Ror2 overexpression in MCF-7 induced a Ror2/Wnt module of the non-canonical Wnt signaling pathway. These targets alter regulation of other pathways involved in cell remodeling processing and cell metabolism. Furthermore, the genes of the Ror2/Wnt module were assessed as a gene signature in patient gene expression data and showed an association with clinical outcome. In summary, results of this study indicate a role of a newly defined Ror2/Wnt module in breast cancer progression and present a link between Ror2 expression and increased cell invasiveness.
Collapse
Affiliation(s)
- Michaela Bayerlová
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Kerstin Menck
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Florian Klemm
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Alexander Wolff
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Tobias Pukrop
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
- Clinic for Internal Medicine III, Hematology and Medical Oncology, University Regensburg, Regensburg, Germany
| | - Claudia Binder
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Tim Beißbarth
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Annalen Bleckmann
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
- Department of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
32
|
Rong X, Zhou Y, Liu Y, Zhao B, Wang B, Wang C, Gong X, Tang P, Lu L, Li Y, Zhao C, Zhou J. Glutathione peroxidase 4 inhibits Wnt/β-catenin signaling and regulates dorsal organizer formation in zebrafish embryos. Development 2017; 144:1687-1697. [PMID: 28302747 DOI: 10.1242/dev.144261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/07/2017] [Indexed: 12/20/2022]
Abstract
The Wnt/β-catenin signaling pathway plays pivotal roles in axis formation during embryogenesis and in adult tissue homeostasis. Glutathione peroxidase 4 (GPX4) is a selenoenzyme and participates in the reduction of peroxides. Its synthesis depends on the availability of the element selenium. However, the roles of GPX4 in vertebrate embryonic development and underlying mechanisms are largely unknown. Here, we show that maternal loss of zebrafish gpx4b promotes embryonic dorsal organizer formation, whereas overexpression of gpx4b inhibits the development of the dorsal organizer. Depletion of human GPX4 and zebrafish gpx4b (GPX4/gpx4b) increases, while GPX4/gpx4b overexpression decreases, Wnt/β-catenin signaling in vivo and in vitro Functional and epistatic studies showed that GPX4 functions at the Tcf/Lef level, independently of selenocysteine activation. Mechanistically, GPX4 interacts with Tcf/Lefs and inhibits Wnt activity by preventing the binding of Tcf/Lefs to the promoters of Wnt target genes, resulting in inhibitory action in the presence of Wnt/β-catenin signaling. Our findings unravel GPX4 as a suppressor of Wnt/β-catenin signals, suggesting a possible relationship between the Wnt/β-catenin pathway and selenium via the association of Tcf/Lef family proteins with GPX4.
Collapse
Affiliation(s)
- Xiaozhi Rong
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.,Institute of Evolution and Marine Biodiversity and College of Marine Biology, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China.,Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Yumei Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Yunzhang Liu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Beibei Zhao
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Bo Wang
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Caixia Wang
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Xiaoxia Gong
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Peipei Tang
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Ling Lu
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Yun Li
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Chengtian Zhao
- Institute of Evolution and Marine Biodiversity and College of Marine Biology, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Jianfeng Zhou
- Key Laboratory of Marine Drugs (Ocean University of China), Chinese Ministry of Education, and School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China .,Laboratory for Marine Drugs and Biological Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| |
Collapse
|
33
|
Abstract
ROR-family receptor tyrosine kinases form a small subfamily of receptor tyrosine kinases (RTKs), characterized by a conserved, unique domain architecture. ROR RTKs are evolutionary conserved throughout the animal kingdom and act as alternative receptors and coreceptors of WNT ligands. The intracellular signaling cascades activated downstream of ROR receptors are diverse, including but not limited to ROR-Frizzled-mediated activation of planar cell polarity signaling, RTK-like signaling, and antagonistic regulation of WNT/β-Catenin signaling. In line with their diverse repertoire of signaling functions, ROR receptors are involved in the regulation of multiple processes in embryonic development such as development of the axial and paraxial mesoderm, the nervous system and the neural crest, the axial and appendicular skeleton, and the kidney. In humans, mutations in the ROR2 gene cause two distinct developmental syndromes, recessive Robinow syndrome (RRS; MIM 268310) and dominant brachydactyly type B1 (BDB1; MIM 113000). In Robinow syndrome patients and animal models, the development of multiple organs is affected, whereas BDB1 results only in shortening of the distal phalanges of fingers and toes, reflecting the diversity of functions and signaling activities of ROR-family RTKs. In this chapter, we give an overview on ROR receptor structure and function. We discuss their signaling functions and role in vertebrate embryonic development with a focus on those developmental processes that are affected by mutations in the ROR2 gene in human patients.
Collapse
|
34
|
Chu CW, Sokol SY. Wnt proteins can direct planar cell polarity in vertebrate ectoderm. eLife 2016; 5. [PMID: 27658614 PMCID: PMC5055393 DOI: 10.7554/elife.16463] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 09/21/2016] [Indexed: 01/10/2023] Open
Abstract
The coordinated orientation of cells across the tissue plane, known as planar cell polarity (PCP), is manifested by the segregation of core PCP proteins to different sides of the cell. Secreted Wnt ligands are involved in many PCP-dependent processes, yet whether they act as polarity cues has been controversial. We show that in Xenopus early ectoderm, the Prickle3/Vangl2 complex was polarized to anterior cell edges and this polarity was disrupted by several Wnt antagonists. In midgastrula embryos, Wnt5a, Wnt11, and Wnt11b, but not Wnt3a, acted across many cell diameters to orient Prickle3/Vangl2 complexes away from their sources regardless of their positions relative to the body axis. The planar polarity of endogenous Vangl2 in the neuroectoderm was similarly redirected by an ectopic Wnt source and disrupted after depletion of Wnt11b in the presumptive posterior region of the embryo. These observations provide evidence for the instructive role of Wnt ligands in vertebrate PCP. DOI:http://dx.doi.org/10.7554/eLife.16463.001
Collapse
Affiliation(s)
- Chih-Wen Chu
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
35
|
|
36
|
Abstract
Extensive molecular characterization of tumors has revealed that the activity of multiple signaling pathways is often simultaneously dampened or enhanced in cancer cells. Aberrant WNT signaling and tyrosine kinase signaling are two pathways that are frequently up- or downregulated in cancer. Although signaling pathways regulated by WNTs, tyrosine kinases, and other factors are often conceptualized as independent entities, the biological reality is likely much more complex. Understanding the mechanisms of crosstalk between multiple signal transduction networks is a key challenge for cancer researchers. The overall goals of this review are to describe mechanisms of crosstalk between WNT and tyrosine kinase pathways in cancer and to discuss how understanding intersections between WNT and tyrosine kinase signaling networks might be exploited to improve current therapies.
Collapse
Affiliation(s)
- Jaimie N Anastas
- Harvard Medical School Department of Cell Biology, Boston, MA; Boston Children's Hospital Division of Newborn Medicine, Boston, MA.
| |
Collapse
|