1
|
Depret N, Gleizes M, Moreau MM, Poirault-Chassac S, Quiedeville A, Carvalho SDS, Venugopal V, Abed ASA, Ezan J, Barthet G, Mulle C, Desmedt A, Marighetto A, Racca C, Montcouquiol M, Sans N. The correct connectivity of the DG-CA3 circuits involved in declarative memory processes depends on Vangl2-dependent planar cell polarity signaling. Prog Neurobiol 2025; 246:102728. [PMID: 39956311 DOI: 10.1016/j.pneurobio.2025.102728] [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: 09/24/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
In the hippocampus, dentate gyrus granule cells connect to CA3 pyramidal cells via their axons, the mossy fibers (Mf). The synaptic terminals of Mfs (Mf boutons, MfBs) form large and complex synapses with thorny excrescences (TE) on the proximal dendrites of CA3 pyramidal cells (PCs). MfB/TE synapses have distinctive "detonator" properties due to low initial release probability and large presynaptic facilitation. The molecular mechanisms shaping the morpho-functional properties of MfB/TE synapses are still poorly understood, though alterations in their morphology are associated with Down syndrome, intellectual disabilities, and Alzheimer's disease. Here, we identify the core PCP gene Vangl2 as essential to the morphogenesis and function of MfB/TE synapses. Vangl2 colocalises with the presynaptic heparan sulfate proteoglycan glypican 4 (GPC4) to stabilise the postsynaptic orphan receptor GPR158. Embryonic loss of Vangl2 disrupts the morphology of MfBs and TEs, impairs ultrastructural and molecular organisation, resulting in defective synaptic transmission and plasticity. In adult, the early loss of Vangl2 results in a number of hippocampus-dependent memory deficits including characteristic flexibility of declarative memory, organisation and retention of working / everyday-like memory. These deficits also lead to abnormal generalisation of memories to salient cues and diminished ability to form detailed contextual memories. Together, these results establish Vangl2 as a key regulator of DG-CA3 connectivity and functions.
Collapse
Affiliation(s)
- Noémie Depret
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Marie Gleizes
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Maïté Marie Moreau
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | | | - Anne Quiedeville
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | | | - Vasika Venugopal
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Alice Shaam Al Abed
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Jérôme Ezan
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Gael Barthet
- Univ. Bordeaux, CNRS, IINS, UMR 5297, Bordeaux F-33000, France
| | | | - Aline Desmedt
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Aline Marighetto
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France
| | - Claudia Racca
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Nathalie Sans
- Univ. Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux F-33000, France.
| |
Collapse
|
2
|
Delmore D, Waghmare I. Molecular evidence supports the functionality of a protein-trapped endogenous allele of Dally-like protein. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001283. [PMID: 39925890 PMCID: PMC11806380 DOI: 10.17912/micropub.biology.001283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/23/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025]
Abstract
The Drosophila glypican Dally-like protein (Dlp) is an evolutionarily-conserved cell-surface protein that modulates extracellular distribution of several secreted ligands for cell signaling. Several fly lines expressing tagged dlp have been used to study the role of Dlp in vivo including the PBac{602.P.SVS-1}dlp [CPTI000445] protein-trap line, which encodes StrepII-Venus-StrepII (SVS)-tagged Dlp from the endogenous locus. dlp is essential for embryonic development, and the SVS-dlp line is homozygous viable. Although this suggests that the SVS-tagged Dlp is functional, it is possible that that the SVS-dlp flies produce wild-type dlp isoform through alternative splicing, contributing to their survival. Here, we used a molecular analysis approach to show that the SVS-dlp flies do not produce wild-type isoform, confirming that the SVS-tagged Dlp is indeed functional.
Collapse
Affiliation(s)
- Drew Delmore
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, United States
| | - Indrayani Waghmare
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, Massachusetts, United States
| |
Collapse
|
3
|
Bruguera ES, Mahoney JP, Weis WI. The co-receptor Tetraspanin12 directly captures Norrin to promote ligand-specific β-catenin signaling. eLife 2025; 13:RP96743. [PMID: 39745873 DOI: 10.7554/elife.96743] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Wnt/β-catenin signaling directs animal development and tissue renewal in a tightly controlled, cell- and tissue-specific manner. In the mammalian central nervous system, the atypical ligand Norrin controls angiogenesis and maintenance of the blood-brain barrier and blood-retina barrier through the Wnt/β-catenin pathway. Like Wnt, Norrin activates signaling by binding and heterodimerizing the receptors Frizzled (Fzd) and low-density lipoprotein receptor-related protein 5 or 6 (LRP5/6), leading to membrane recruitment of the intracellular transducer Dishevelled (Dvl) and ultimately stabilizing the transcriptional coactivator β-catenin. Unlike Wnt, the cystine knot ligand Norrin only signals through Fzd4 and additionally requires the co-receptor Tetraspanin12 (Tspan12); however, the mechanism underlying Tspan12-mediated signal enhancement is unclear. It has been proposed that Tspan12 integrates into the Norrin-Fzd4 complex to enhance Norrin-Fzd4 affinity or otherwise allosterically modulate Fzd4 signaling. Here, we measure direct, high-affinity binding between purified Norrin and Tspan12 in a lipid environment and use AlphaFold models to interrogate this interaction interface. We find that Tspan12 and Fzd4 can simultaneously bind Norrin and that a pre-formed Tspan12/Fzd4 heterodimer, as well as cells co-expressing Tspan12 and Fzd4, more efficiently capture low concentrations of Norrin than Fzd4 alone. We also show that Tspan12 competes with both heparan sulfate proteoglycans and LRP6 for Norrin binding and that Tspan12 does not impact Fzd4-Dvl affinity in the presence or absence of Norrin. Our findings suggest that Tspan12 does not allosterically enhance Fzd4 binding to Norrin or Dvl, but instead functions to directly capture Norrin upstream of signaling.
Collapse
Affiliation(s)
- Elise S Bruguera
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, United States
| | - Jacob P Mahoney
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, United States
| | - William I Weis
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
4
|
Bruguera ES, Mahoney JP, Weis WI. The co-receptor Tetraspanin12 directly captures Norrin to promote ligand-specific β-catenin signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.03.578714. [PMID: 38352533 PMCID: PMC10862866 DOI: 10.1101/2024.02.03.578714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Wnt/β-catenin signaling directs animal development and tissue renewal in a tightly controlled, cell- and tissue-specific manner. In the mammalian central nervous system, the atypical ligand Norrin controls angiogenesis and maintenance of the blood-brain barrier and blood-retina barrier through the Wnt/β-catenin pathway. Like Wnt, Norrin activates signaling by binding and heterodimerizing the receptors Frizzled (Fzd) and Low-density lipoprotein receptor-related protein 5 or 6 (LRP5/6), leading to membrane recruitment of the intracellular transducer Dishevelled (Dvl) and ultimately stabilizing the transcriptional coactivator β-catenin. Unlike Wnt, the cystine-knot ligand Norrin only signals through Fzd4 and additionally requires the co-receptor Tetraspanin12 (Tspan12); however, the mechanism underlying Tspan12-mediated signal enhancement is unclear. It has been proposed that Tspan12 integrates into the Norrin-Fzd4 complex to enhance Norrin-Fzd4 affinity or otherwise allosterically modulate Fzd4 signaling. Here, we measure direct, high-affinity binding between purified Norrin and Tspan12 in a lipid environment and use AlphaFold models to interrogate this interaction interface. We find that Tspan12 and Fzd4 can simultaneously bind Norrin and that a pre-formed Tspan12/Fzd4 heterodimer, as well as cells co-expressing Tspan12 and Fzd4, more efficiently capture low concentrations of Norrin than Fzd4 alone. We also show that Tspan12 competes with both heparan sulfate proteoglycans and LRP6 for Norrin binding and that Tspan12 does not impact Fzd4-Dvl affinity in the presence or absence of Norrin. Our findings suggest that Tspan12 does not allosterically enhance Fzd4 binding to Norrin or Dvl, but instead functions to directly capture Norrin upstream of signaling.
Collapse
Affiliation(s)
- Elise S Bruguera
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Jacob P Mahoney
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - William I Weis
- Departments of Molecular & Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
5
|
Ell CM, Safyan A, Chayengia M, Kustermann MMM, Lorenz J, Schächtle M, Pyrowolakis G. A genome-engineered tool set for Drosophila TGF-β/BMP signaling studies. Development 2024; 151:dev204222. [PMID: 39494616 DOI: 10.1242/dev.204222] [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: 07/01/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
Ligands of the TGF-β/BMP superfamily are crucially involved in the regulation of growth, patterning and organogenesis and can act as long-range morphogens. Essential for understanding TGF-β/BMP signaling dynamics and regulation are tools that allow monitoring and manipulating pathway components at physiological expression levels and endogenous spatiotemporal patterns. We used genome engineering to generate a comprehensive library of endogenously epitope- or fluorescent-tagged versions of receptors, co-receptors, transcription factors and key feedback regulators of the Drosophila BMP and Activin signaling pathways. We demonstrate that the generated alleles are biologically active and can be used for assessing tissue and subcellular distribution of the corresponding proteins. Furthermore, we show that the genomic platforms can be used for in locus structure-function and cis-regulatory analyses. Finally, we present a complementary set of protein binder-based tools, which allow visualization as well as manipulation of the stability and subcellular localization of epitope-tagged proteins, providing new tools for the analysis of BMP signaling and beyond.
Collapse
Affiliation(s)
- Clara-Maria Ell
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - Abu Safyan
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
- International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Mrinal Chayengia
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - Manuela M M Kustermann
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - Jennifer Lorenz
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - Melanie Schächtle
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| | - George Pyrowolakis
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, HMH, Habsburgerstr. 49, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
6
|
Manouchehri JM, Datta J, Marcho LM, Reardon JJ, Stover D, Wesolowski R, Borate U, Cheng TYD, Schnell PM, Ramaswamy B, Sizemore GM, Rubinstein MP, Cherian MA. The role of heparan sulfate in enhancing the chemotherapeutic response in triple-negative breast cancer. Breast Cancer Res 2024; 26:153. [PMID: 39506780 PMCID: PMC11539583 DOI: 10.1186/s13058-024-01906-6] [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: 04/17/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Breast cancer, one of the most common forms of cancer, is associated with the highest cancer-related mortality among women worldwide. In comparison to other types of breast cancer, patients diagnosed with the triple-negative breast cancer (TNBC) subtype have the worst outcome because current therapies do not produce long-lasting responses. Hence, innovative therapies that produce persisting responses are a critical need. We previously discovered that hyperactivating purinergic receptors (P2RXs) by increasing extracellular adenosine triphosphate (eATP) concentrations enhances TNBC cell lines' response to chemotherapy. Heparan sulfate inhibits multiple extracellular ATPases, so it is a molecule of interest in this regard. In turn, heparanase degrades polysulfated polysaccharide heparan sulfate. Importantly, previous work suggests that breast cancer and other cancers express heparanase at high levels. Hence, as heparan sulfate can inhibit extracellular ATPases to facilitate eATP accumulation, it may intensify responses to chemotherapy. We postulated that heparanase inhibitors would exacerbate chemotherapy-induced decreases in TNBC cell viability by increasing heparan sulfate in the cellular microenvironment and hence, augmenting eATP. METHODS We treated TNBC cell lines MDA-MB 231, Hs 578t, and MDA-MB 468 and non-tumorigenic immortal mammary epithelial MCF-10A cells with paclitaxel (cytotoxic chemotherapeutic) with or without the heparanase inhibitor OGT 2115 and/or supplemental heparan sulfate. We evaluated cell viability and the release of eATP. Also, we compared the expression of heparanase protein in cell lines and tissues by immunoblot and immunohistochemistry, respectively. In addition, we examined breast-cancer-initiating cell populations using tumorsphere formation efficiency assays on treated cells. RESULTS We found that combining heparanase inhibitor OGT 2115 with chemotherapy decreased TNBC cell viability and tumorsphere formation through increases in eATP and activation of purinergic receptors as compared to TNBC cells treated with single-agent paclitaxel. CONCLUSION Our data shows that by preventing heparan sulfate breakdown, heparanase inhibitors make TNBC cells more susceptible to chemotherapy by enhancing eATP concentrations.
Collapse
Affiliation(s)
- Jasmine M Manouchehri
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Jharna Datta
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Lynn M Marcho
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Jesse J Reardon
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Daniel Stover
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Uma Borate
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Ting-Yuan David Cheng
- Division of Cancer Prevention and Control, Department of Internal Medicine, The Ohio State University, Suite 525, 1590 North High St., Columbus, OH, 43201, USA
| | - Patrick M Schnell
- Division of Biostatistics, The Ohio State University College of Public Health, 1841 Neil Ave., Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Gina M Sizemore
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Mark P Rubinstein
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Mathew A Cherian
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA.
- Division of Medical Oncology, 460 W 12th Ave., 888 BRT, Columbus, OH, 43210, USA.
| |
Collapse
|
7
|
Gao Y, Feng J, Zhang Y, Yi M, Zhang L, Yan Y, Zhu AJ, Liu M. Ehbp1 orchestrates orderly sorting of Wnt/Wingless to the basolateral and apical cell membranes. EMBO Rep 2024; 25:5053-5079. [PMID: 39402333 PMCID: PMC11549480 DOI: 10.1038/s44319-024-00289-1] [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: 04/15/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Wingless (Wg)/Wnt signaling plays a critical role in both development and adult tissue homeostasis. In the Drosophila larval wing disc epithelium, the orderly delivery of Wg/Wnt to the apical and basal cell surfaces is essential for wing development. Here, we identified Ehbp1 as the switch that dictates the direction of Wg/Wnt polarized intracellular transport: the Adaptor Protein complex 1 (AP-1) delivers Wg/Wnt to the basolateral cell surface, and its sequestration by Ehbp1 redirects Wg/Wnt for apical delivery. Genetic analyses showed that Ehbp1 specifically regulates the polarized distribution of Wg/Wnt, a process that depends on the dedicated Wg/Wnt cargo receptor Wntless. Mechanistically, Ehbp1 competes with Wntless for AP-1 binding, thereby preventing the unregulated basolateral Wg/Wnt transport. Reducing Ehbp1 expression, or removing the coiled-coil motifs within its bMERB domain, leads to basolateral Wg/Wnt accumulation. Importantly, the regulation of polarized Wnt delivery by EHBP1 is conserved in vertebrates. The generality of this switch mechanism for regulating intracellular transport remains to be determined in future studies.
Collapse
Affiliation(s)
- Yuan Gao
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jing Feng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yansong Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, 610213, China
| | - Mengyuan Yi
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Lebing Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yan Yan
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Alan Jian Zhu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, 610213, China.
| | - Min Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
| |
Collapse
|
8
|
Zhang X, Wang Y, Zhao W, Yang S, Moussian B, Zhao Z, Zhang J, Dong W. Excess Dally-like Induces Malformation of Drosophila Legs. Cells 2024; 13:1199. [PMID: 39056781 PMCID: PMC11274743 DOI: 10.3390/cells13141199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Glypicans are closely associated with organ development and tumorigenesis in animals. Dally-like (Dlp), a membrane-bound glypican, plays pivotal roles in various biological processes in Drosophila. In this study, we observed that an excess of Dlp led to the malformation of legs, particularly affecting the distal part. Accordingly, the leg disc was shrunken and frequently exhibited aberrant morphology. In addition, elevated Dlp levels induced ectopic cell death with no apparent cell proliferation changes. Furthermore, Dlp overexpression in the posterior compartment significantly altered Wingless (Wg) distribution. We observed a marked expansion of Wg distribution within the posterior compartment, accompanied by a corresponding decrease in the anterior compartment. It appears that excess Dlp guides Wg to diffuse to cells with higher Dlp levels. In addition, the distal-less (dll) gene, which is crucial for leg patterning, was up-regulated significantly. Notably, dachshund (dac) and homothorax (hth) expression, also essential for leg patterning and development, only appeared to be negligibly affected. Based on these findings, we speculate that excess Dlp may contribute to malformations of the distal leg region of Drosophila, possibly through its influence on Wg distribution, dll expression and induced cell death. Our research advances the understanding of Dlp function in Drosophila leg development.
Collapse
Affiliation(s)
- Xubo Zhang
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Yi Wang
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Wenting Zhao
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Shumin Yang
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Bernard Moussian
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Centre National de la Recherche Scientifique, Institut Sophia Agrobiotech, Sophia Antipolis, Université Côte d′Azur, 06108 Nice, France
| | - Zhangwu Zhao
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Jianzhen Zhang
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| | - Wei Dong
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
9
|
Jiménez-Jiménez C, Grobe K, Guerrero I. Hedgehog on the Move: Glypican-Regulated Transport and Gradient Formation in Drosophila. Cells 2024; 13:418. [PMID: 38474382 PMCID: PMC10930589 DOI: 10.3390/cells13050418] [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: 01/30/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Glypicans (Glps) are a family of heparan sulphate proteoglycans that are attached to the outer plasma membrane leaflet of the producing cell by a glycosylphosphatidylinositol anchor. Glps are involved in the regulation of many signalling pathways, including those that regulate the activities of Wnts, Hedgehog (Hh), Fibroblast Growth Factors (FGFs), and Bone Morphogenetic Proteins (BMPs), among others. In the Hh-signalling pathway, Glps have been shown to be essential for ligand transport and the formation of Hh gradients over long distances, for the maintenance of Hh levels in the extracellular matrix, and for unimpaired ligand reception in distant recipient cells. Recently, two mechanistic models have been proposed to explain how Hh can form the signalling gradient and how Glps may contribute to it. In this review, we describe the structure, biochemistry, and metabolism of Glps and their interactions with different components of the Hh-signalling pathway that are important for the release, transport, and reception of Hh.
Collapse
Affiliation(s)
- Carlos Jiménez-Jiménez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Nicolás Cabrera 1, E-28049 Madrid, Spain;
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| | - Isabel Guerrero
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Nicolás Cabrera 1, E-28049 Madrid, Spain;
| |
Collapse
|
10
|
Deshpande G, Ng C, Jourjine N, Chiew JW, Dasilva J, Schedl P. Hedgehog signaling guides migration of primordial germ cells to the Drosophila somatic gonad. Genetics 2023; 225:iyad165. [PMID: 37708366 PMCID: PMC10627259 DOI: 10.1093/genetics/iyad165] [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: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023] Open
Abstract
In addition to inducing nonautonomous specification of cell fate in both Drosophila and vertebrates, the Hedgehog pathway guides cell migration in a variety of different tissues. Although its role in axon guidance in the vertebrate nervous system is widely recognized, its role in guiding the migratory path of primordial germ cells (PGCs) from the outside surface of the Drosophila embryo through the midgut and mesoderm to the SGPs (somatic gonadal precursors) has been controversial. Here we present new experiments demonstrating (1) that Hh produced by mesodermal cells guides PGC migration, (2) that HMG CoenzymeA reductase (Hmgcr) potentiates guidance signals emanating from the SGPs, functioning upstream of hh and of 2 Hh pathway genes important for Hh-containing cytonemes, and (3) that factors required in Hh receiving cells in other contexts function in PGCs to help direct migration toward the SGPs. We also compare the data reported by 4 different laboratories that have studied the role of the Hh pathway in guiding PGC migration.
Collapse
Affiliation(s)
- Girish Deshpande
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Chris Ng
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Nicholas Jourjine
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Joy Wan Chiew
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Juliana Dasilva
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Paul Schedl
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
11
|
Dong Z, Pang L, Liu Z, Sheng Y, Li X, Thibault X, Reilein A, Kalderon D, Huang J. Single-cell expression profile of Drosophila ovarian follicle stem cells illuminates spatial differentiation in the germarium. BMC Biol 2023; 21:143. [PMID: 37340484 PMCID: PMC10283321 DOI: 10.1186/s12915-023-01636-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND How stem cell populations are organized and regulated within adult tissues is important for understanding cancer origins and for developing cell replacement strategies. Paradigms such as mammalian gut stem cells and Drosophila ovarian follicle stem cells (FSC) are characterized by population asymmetry, in which stem cell division and differentiation are separately regulated processes. These stem cells behave stochastically regarding their contributions to derivative cells and also exhibit dynamic spatial heterogeneity. Drosophila FSCs provide an excellent model for understanding how a community of active stem cells maintained by population asymmetry is regulated. Here, we use single-cell RNA sequencing to profile the gene expression patterns of FSCs and their immediate derivatives to investigate heterogeneity within the stem cell population and changes associated with differentiation. RESULTS We describe single-cell RNA sequencing studies of a pre-sorted population of cells that include FSCs and the neighboring cell types, escort cells (ECs) and follicle cells (FCs), which they support. Cell-type assignment relies on anterior-posterior (AP) location within the germarium. We clarify the previously determined location of FSCs and use spatially targeted lineage studies as further confirmation. The scRNA profiles among four clusters are consistent with an AP progression from anterior ECs through posterior ECs and then FSCs, to early FCs. The relative proportion of EC and FSC clusters are in good agreement with the prevalence of those cell types in a germarium. Several genes with graded profiles from ECs to FCs are highlighted as candidate effectors of the inverse gradients of the two principal signaling pathways, Wnt and JAK-STAT, that guide FSC differentiation and division. CONCLUSIONS Our data establishes an important resource of scRNA-seq profiles for FSCs and their immediate derivatives that is based on precise spatial location and functionally established stem cell identity, and facilitates future genetic investigation of regulatory interactions guiding FSC behavior.
Collapse
Affiliation(s)
- Zhi Dong
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Lan Pang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Zhiguo Liu
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Yifeng Sheng
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China
| | - Xavier Thibault
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Amy Reilein
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| | - Jianhua Huang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
12
|
Iyer KS, Prabhakara C, Mayor S, Rao M. Cellular compartmentalisation and receptor promiscuity as a strategy for accurate and robust inference of position during morphogenesis. eLife 2023; 12:e79257. [PMID: 36877545 PMCID: PMC9988261 DOI: 10.7554/elife.79257] [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: 04/05/2022] [Accepted: 01/14/2023] [Indexed: 03/07/2023] Open
Abstract
Precise spatial patterning of cell fate during morphogenesis requires accurate inference of cellular position. In making such inferences from morphogen profiles, cells must contend with inherent stochasticity in morphogen production, transport, sensing and signalling. Motivated by the multitude of signalling mechanisms in various developmental contexts, we show how cells may utilise multiple tiers of processing (compartmentalisation) and parallel branches (multiple receptor types), together with feedback control, to bring about fidelity in morphogenetic decoding of their positions within a developing tissue. By simultaneously deploying specific and nonspecific receptors, cells achieve a more accurate and robust inference. We explore these ideas in the patterning of Drosophila melanogaster wing imaginal disc by Wingless morphogen signalling, where multiple endocytic pathways participate in decoding the morphogen gradient. The geometry of the inference landscape in the high dimensional space of parameters provides a measure for robustness and delineates stiff and sloppy directions. This distributed information processing at the scale of the cell highlights how local cell autonomous control facilitates global tissue scale design.
Collapse
Affiliation(s)
- Krishnan S Iyer
- Simons Center for the Study of Living Machines, National Center for Biological Sciences - TIFRBangaloreIndia
| | | | - Satyajit Mayor
- National Center for Biological Sciences - TIFRBangaloreIndia
| | - Madan Rao
- Simons Center for the Study of Living Machines, National Center for Biological Sciences - TIFRBangaloreIndia
| |
Collapse
|
13
|
Abstract
Wnts are secreted proteins that control stem cell maintenance, cell fate decisions, and growth during development and adult homeostasis. Wnts carry a post-translational modification not seen in any other secreted protein: during biosynthesis, they are appended with a palmitoleoyl moiety that is required for signaling but also impairs solubility and hence diffusion in the extracellular space. In some contexts, Wnts act only in a juxtacrine manner but there are also instances of long range action. Several proteins and processes ensure that active Wnts reach the appropriate target cells. Some, like Porcupine, Wntless, and Notum are dedicated to Wnt function; we describe their activities in molecular detail. We also outline how the cell infrastructure (secretory, endocytic, and retromer pathways) contribute to the progression of Wnts from production to delivery. We then address how Wnts spread in the extracellular space and form a signaling gradient despite carrying a hydrophobic moiety. We highlight particularly the role of lipid-binding Wnt interactors and heparan sulfate proteoglycans. Finally, we briefly discuss how evolution might have led to the emergence of this unusual signaling pathway.
Collapse
|
14
|
Otaki JM, Nakazato Y. Butterfly Wing Color Pattern Modification Inducers May Act on Chitin in the Apical Extracellular Site: Implications in Morphogenic Signals for Color Pattern Determination. BIOLOGY 2022; 11:1620. [PMID: 36358322 PMCID: PMC9687432 DOI: 10.3390/biology11111620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 09/28/2023]
Abstract
Butterfly wing color patterns are modified by various treatments, such as temperature shock, injection of chemical inducers, and covering materials on pupal wing tissue. Their mechanisms of action have been enigmatic. Here, we investigated the mechanisms of color pattern modifications usingthe blue pansy butterfly Junoniaorithya. We hypothesized that these modification-inducing treatments act on the pupal cuticle or extracellular matrix (ECM). Mechanical load tests revealed that pupae treated with cold shock or chemical inducers were significantly less rigid, suggesting that these treatments made cuticle formation less efficient. A known chitin inhibitor, FB28 (fluorescent brightener 28), was discovered to efficiently induce modifications. Taking advantage of its fluorescent character, fluorescent signals from FB28 were observed in live pupae in vivo from the apical extracellular side and were concentrated at the pupal cuticle focal spots immediately above the eyespot organizing centers. It was shown that chemical modification inducers and covering materials worked additively. Taken together, various modification-inducing treatments likely act extracellularly on chitin or other polysaccharides to inhibit pupal cuticle formation or ECM function, which probably causes retardation of morphogenic signals. It is likely that an interactive ECM is required for morphogenic signals for color pattern determination to travel long distances.
Collapse
Affiliation(s)
- Joji M. Otaki
- The BCPH Unit of Molecular Physiology, Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Okinawa 903-0213, Japan
| | | |
Collapse
|
15
|
Hayashida K, Aquino RS, Park PW. Coreceptor Functions of Cell Surface Heparan Sulfate Proteoglycans. Am J Physiol Cell Physiol 2022; 322:C896-C912. [PMID: 35319900 PMCID: PMC9109798 DOI: 10.1152/ajpcell.00050.2022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Receptor-ligand interactions play an important role in many biological processes by triggering specific cellular responses. These interactions are frequently regulated by coreceptors that facilitate, alter, or inhibit signaling. Coreceptors work in parallel with other specific and accessory molecules to coordinate receptor-ligand interactions. Cell surface heparan sulfate proteoglycans (HSPGs) function as unique coreceptors because they can bind to many ligands and receptors through their HS and core protein motifs. Cell surface HSPGs are typically expressed in abundance of the signaling receptors and, thus, are capable of mediating the initial binding of ligands to the cell surface. HSPG coreceptors do not possess kinase domains or intrinsic enzyme activities and, for the most part, binding to cell surface HSPGs does not directly stimulate intracellular signaling. Because of these features, cell surface HSPGs primarily function as coreceptors for many receptor-ligand interactions. Given that cell surface HSPGs are widely conserved, they likely serve fundamental functions to preserve basic physiological processes. Indeed, cell surface HSPGs can support specific cellular interactions with growth factors, morphogens, chemokines, extracellular matrix (ECM) components, and microbial pathogens and their secreted virulence factors. Through these interactions, HSPG coreceptors regulate cell adhesion, proliferation, migration and differentiation, and impact the onset, progression, and outcome of pathophysiological processes, such as development, tissue repair, inflammation, infection, and tumorigenesis. This review seeks to provide an overview of the various mechanisms of how cell surface HSPGs function as coreceptors.
Collapse
Affiliation(s)
- Kazutaka Hayashida
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Rafael S Aquino
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Pyong Woo Park
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Liu YC, Wierbowski BM, Salic A. Hedgehog pathway modulation by glypican 3-conjugated heparan sulfate. J Cell Sci 2022; 135:274739. [PMID: 35142364 PMCID: PMC8977055 DOI: 10.1242/jcs.259297] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 02/04/2022] [Indexed: 11/20/2022] Open
Abstract
Glypicans are a family of cell surface heparan sulfate proteoglycans that play critical roles in multiple cell signaling pathways. Glypicans consist of a globular core, an unstructured stalk modified with sulfated glycosaminoglycan chains, and a glycosylphosphatidylinositol anchor. Though these structural features are conserved, their individual contribution to glypican function remains obscure. Here, we investigate how glypican 3 (GPC3), which is mutated in Simpson-Golabi-Behmel tissue overgrowth syndrome, regulates Hedgehog signaling. We find that GPC3 is necessary for the Hedgehog response, surprisingly controlling a downstream signal transduction step. Purified GPC3 ectodomain rescues signaling when artificially recruited to the surface of GPC3-deficient cells but has dominant-negative activity when unattached. Strikingly, the purified stalk, modified with heparan sulfate but not chondroitin sulfate, is necessary and sufficient for activity. Our results demonstrate a novel function for GPC3-associated heparan sulfate and provide a framework for the functional dissection of glycosaminoglycans by in vivo biochemical complementation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Yulu Cherry Liu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.,Department of Biology, Hood College, Frederick, MD 21701, USA
| | | | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Abstract
The Drosophila wing imaginal disc is a tissue of undifferentiated cells that are precursors of the wing and most of the notum of the adult fly. The wing disc first forms during embryogenesis from a cluster of ∼30 cells located in the second thoracic segment, which invaginate to form a sac-like structure. They undergo extensive proliferation during larval stages to form a mature larval wing disc of ∼35,000 cells. During this time, distinct cell fates are assigned to different regions, and the wing disc develops a complex morphology. Finally, during pupal stages the wing disc undergoes morphogenetic processes and then differentiates to form the adult wing and notum. While the bulk of the wing disc comprises epithelial cells, it also includes neurons and glia, and is associated with tracheal cells and muscle precursor cells. The relative simplicity and accessibility of the wing disc, combined with the wealth of genetic tools available in Drosophila, have combined to make it a premier system for identifying genes and deciphering systems that play crucial roles in animal development. Studies in wing imaginal discs have made key contributions to many areas of biology, including tissue patterning, signal transduction, growth control, regeneration, planar cell polarity, morphogenesis, and tissue mechanics.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
18
|
Waghmare I, Page-McCaw A. Regulation of Wnt distribution and function by Drosophila glypicans. J Cell Sci 2022; 135:274233. [PMID: 35112708 PMCID: PMC8918805 DOI: 10.1242/jcs.259405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The extracellular distribution of secreted Wnt proteins is crucial for their ability to induce a response in target cells at short and long ranges to ensure proper development. Wnt proteins are evolutionarily conserved ligands that are lipid-modified, and their hydrophobic nature interferes with their solubility in the hydrophilic extracellular environment. This raises the question of how Wnt proteins spread extracellularly despite their lipid modifications, which are essential for both their secretion and function. Seminal studies on Drosophila Wingless (Wg), a prototypical Wnt, have discovered multiple mechanisms by which Wnt proteins spread. A central theme emerges from these studies: the Wnt lipid moiety is shielded from the aqueous environment, allowing the ligands to spread and remain viable for signaling. Wnt distribution in vivo is primarily facilitated by glypicans, which are cell-surface heparan sulfate proteoglycans, and recent studies have further provided mechanistic insight into how glypicans facilitate Wnt distribution. In this Review, we discuss the many diverse mechanisms of Wnt distribution, with a particular focus on glypican-mediated mechanisms.
Collapse
|
19
|
Brown MA, Ried T. Shifting the Focus of Signaling Abnormalities in Colon Cancer. Cancers (Basel) 2022; 14:784. [PMID: 35159051 PMCID: PMC8834070 DOI: 10.3390/cancers14030784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/26/2022] [Accepted: 01/30/2022] [Indexed: 12/12/2022] Open
Abstract
Colon cancer tumorigenesis occurs incrementally. The process involves the acquisition of mutations which typically follow an established pattern: activation of WNT signaling, activation of RAS signaling, and inhibition of TGF-β signaling. This arrangement recapitulates, to some degree, the stem cell niche of the intestinal epithelium, which maintains WNT and EGF activity while suppressing TGF-β. The resemblance between the intestinal stem cell environment and colon cancer suggests that the concerted activity of these pathways generates and maintains a potent growth-inducing stimulus. However, each pathway has a myriad of downstream targets, making it difficult to identify which aspects of these pathways are drivers. To address this, we utilize the cell cycle, the ultimate regulator of cell proliferation, as a foundation for cross-pathway integration. We attempt to generate an overview of colon cancer signaling patterns by integrating the major colon cancer signaling pathways in the context of cell replication, specifically, the entrance from G1 into S-phase.
Collapse
Affiliation(s)
| | - Thomas Ried
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA;
| |
Collapse
|
20
|
An itch for things remote: The journey of Wnts. Curr Top Dev Biol 2022; 150:91-128. [DOI: 10.1016/bs.ctdb.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
21
|
Molecular Genetic Techniques for the Proteoglycan Functions in Drosophila. Methods Mol Biol 2021. [PMID: 34626396 DOI: 10.1007/978-1-0716-1398-6_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Several classes of heparan sulfate proteoglycan (HSPG) core proteins and all HS biosynthetic/modifying enzymes are evolutionarily conserved from human to Drosophila melanogaster. This genetically tractable model offers highly sophisticated techniques to manipulate gene function in a spatially and temporally controlled manner. Thus, Drosophila genetics has been a powerful system to explore functions of HSPGs in vivo. In this chapter, we will introduce three genetic techniques available in Drosophila: TARGET (temporal and regional gene expression targeting), MARCM (mosaic analysis with a repressible cell marker), and FLP-Out.
Collapse
|
22
|
Zaykov V, Chaqour B. The CCN2/CTGF interactome: an approach to understanding the versatility of CCN2/CTGF molecular activities. J Cell Commun Signal 2021; 15:567-580. [PMID: 34613590 DOI: 10.1007/s12079-021-00650-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/23/2021] [Indexed: 01/16/2023] Open
Abstract
Cellular communication network 2 (CCN2), also known as connective tissue growth factor (CTGF) regulates diverse cellular processes, some at odds with others, including adhesion, proliferation, apoptosis, and extracellular matrix (ECM) protein synthesis. Although a cause-and-effect relationship between CCN2/CTGF expression and local fibrotic reactions has initially been established, CCN2/CTGF manifests cell-, tissue-, and context-specific functions and differentially affects developmental and pathological processes ranging from progenitor cell fate decisions and angiogenesis to inflammation and tumorigenesis. CCN2/CTGF multimodular structure, binding to and activation or inhibition of multiple cell surface receptors, growth factors and ECM proteins, and susceptibility for proteolytic cleavage highlight the complexity to CCN2/CTGF biochemical attributes. CCN2/CTGF expression and dosage in the local environment affects a defined community of its interacting partners, and this results in sequestration of growth factors, interference with or potentiation of ligand-receptor binding, cellular internalization of CCN2/CTGF, inhibition or activation of proteases, and generation of CCN2/CTGF degradome products that add molecular diversity and expand the repertoire of functional modules in the cells and their microenvironment. Through these interactions, different intracellular signals and cellular responses are elicited culminating into physiological or pathological reactions. Thus, the CCN2/CTGF interactome is a defining factor of its tissue- and context-specific effects. Mapping of new CCN2/CTGF binding partners might shed light on yet unknown roles of CCN2/CTGF and provide a solid basis for tissue-specific targeting this molecule or its interacting partners in a therapeutic context.
Collapse
Affiliation(s)
- Viktor Zaykov
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA
| | - Brahim Chaqour
- Department of Cell Biology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA. .,Department of Ophthalmology, State University of New York (SUNY), Downstate Health Science University, 450 Clarkson Avenue, MSC 5, Brooklyn, NY, 11203, USA.
| |
Collapse
|
23
|
Mehta S, Hingole S, Chaudhary V. The Emerging Mechanisms of Wnt Secretion and Signaling in Development. Front Cell Dev Biol 2021; 9:714746. [PMID: 34485301 PMCID: PMC8415634 DOI: 10.3389/fcell.2021.714746] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/20/2021] [Indexed: 12/22/2022] Open
Abstract
Wnts are highly-conserved lipid-modified secreted proteins that activate multiple signaling pathways. These pathways regulate crucial processes during various stages of development and maintain tissue homeostasis in adults. One of the most fascinating aspects of Wnt protein is that despite being hydrophobic, they are known to travel several cell distances in the extracellular space. Research on Wnts in the past four decades has identified several factors and uncovered mechanisms regulating their expression, secretion, and mode of extracellular travel. More recently, analyses on the importance of Wnt protein gradients in the growth and patterning of developing tissues have recognized the complex interplay of signaling mechanisms that help in maintaining tissue homeostasis. This review aims to present an overview of the evidence for the various modes of Wnt protein secretion and signaling and discuss mechanisms providing precision and robustness to the developing tissues.
Collapse
Affiliation(s)
| | | | - Varun Chaudhary
- Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, India
| |
Collapse
|
24
|
Simon E, Jiménez-Jiménez C, Seijo-Barandiarán I, Aguilar G, Sánchez-Hernández D, Aguirre-Tamaral A, González-Méndez L, Ripoll P, Guerrero I. Glypicans define unique roles for the Hedgehog co-receptors boi and ihog in cytoneme-mediated gradient formation. eLife 2021; 10:64581. [PMID: 34355694 PMCID: PMC8410076 DOI: 10.7554/elife.64581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
The conserved family of Hedgehog (Hh) signaling proteins plays a key role in cell–cell communication in development, tissue repair, and cancer progression, inducing distinct concentration-dependent responses in target cells located at short and long distances. One simple mechanism for long distance dispersal of the lipid modified Hh is the direct contact between cell membranes through filopodia-like structures known as cytonemes. Here we have analyzed in Drosophila the interaction between the glypicans Dally and Dally-like protein, necessary for Hh signaling, and the adhesion molecules and Hh coreceptors Ihog and Boi. We describe that glypicans are required to maintain the levels of Ihog, but not of Boi. We also show that the overexpression of Ihog, but not of Boi, regulates cytoneme dynamics through their interaction with glypicans, the Ihog fibronectin III domains being essential for this interaction. Our data suggest that the regulation of glypicans over Hh signaling is specifically given by their interaction with Ihog in cytonemes. Contrary to previous data, we also show that there is no redundancy of Ihog and Boi functions in Hh gradient formation, being Ihog, but not of Boi, essential for the long-range gradient.
Collapse
Affiliation(s)
- Eléanor Simon
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Carlos Jiménez-Jiménez
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Irene Seijo-Barandiarán
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Gustavo Aguilar
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain.,Growth and Development, University of Basel, Biozentrum, Switzerland
| | - David Sánchez-Hernández
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Adrián Aguirre-Tamaral
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Laura González-Méndez
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Pedro Ripoll
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Isabel Guerrero
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| |
Collapse
|
25
|
Salbach-Hirsch J, Rauner M, Hofbauer C, Hofbauer LC. New insights into the role of glycosaminoglycans in the endosteal bone microenvironment. Biol Chem 2021; 402:1415-1425. [PMID: 34323057 DOI: 10.1515/hsz-2021-0174] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/14/2021] [Indexed: 12/19/2022]
Abstract
The bone microenvironment is a complex tissue in which heterogeneous cell populations of hematopoietic and mesenchymal origin interact with environmental cues to maintain tissue integrity. Both cellular and matrix components are subject to physiologic challenges and can dynamically respond by modifying cell/matrix interactions. When either component is impaired, the physiologic balance is lost. Here, we review the current state of knowledge of how glycosaminoglycans - organic components of the bone extracellular matrix - influence the bone micromilieu. We point out how they interact with mediators of distinct signaling pathways such as the RANKL/OPG axis, BMP and WNT signaling, and affect the activity of bone remodeling cells within the endosteal niche summarizing their potential for therapeutic intervention.
Collapse
Affiliation(s)
- Juliane Salbach-Hirsch
- Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Medical Center, Technische Universität Dresden, D-01307 Dresden, Germany
- Center for Healthy Aging, Medical Center, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Martina Rauner
- Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Medical Center, Technische Universität Dresden, D-01307 Dresden, Germany
- Center for Healthy Aging, Medical Center, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Christine Hofbauer
- NCT Dresden and University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Lorenz C Hofbauer
- Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Medical Center, Technische Universität Dresden, D-01307 Dresden, Germany
- Center for Healthy Aging, Medical Center, Technische Universität Dresden, D-01307 Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), D-01307 Dresden, Germany
| |
Collapse
|
26
|
Wang X, LaFever KS, Waghmare I, Page-McCaw A. Extracellular spreading of Wingless is required for Drosophila oogenesis. PLoS Genet 2021; 17:e1009469. [PMID: 33798197 PMCID: PMC8046344 DOI: 10.1371/journal.pgen.1009469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/14/2021] [Accepted: 03/05/2021] [Indexed: 11/18/2022] Open
Abstract
Recent studies have investigated whether the Wnt family of extracellular ligands can signal at long range, spreading from their source and acting as morphogens, or whether they signal only in a juxtacrine manner to neighboring cells. The original evidence for long-range Wnt signaling arose from studies of Wg, a Drosophila Wnt protein, which patterns the wing disc over several cell diameters from a central source of Wg ligand. However, the requirement of long-range Wg for patterning was called into question when it was reported that replacing the secreted protein Wg with a membrane-tethered version, NRT-Wg, results in flies with normally patterned wings. We and others previously reported that Wg spreads in the ovary about 50 μm or 5 cell diameters, from the cap cells to the follicle stem cells (FSCs) and that Wg stimulates FSC proliferation. We used the NRT-wg flies to analyze the consequence of tethering Wg to the cap cells. NRT-wg homozygous flies are sickly, but we found that hemizygous NRT-wg/null flies, carrying only one copy of tethered Wingless, were significantly healthier. Despite their overall improved health, these hemizygous flies displayed dramatic reductions in fertility and in FSC proliferation. Further, FSC proliferation was nearly undetectable when the wg locus was converted to NRT-wg only in adults, and the resulting germarium phenotype was consistent with a previously reported wg loss-of-function phenotype. We conclude that Wg protein spreads from its source cells in the germarium to promote FSC proliferation.
Collapse
Affiliation(s)
- Xiaoxi Wang
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Kimberly S. LaFever
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Indrayani Waghmare
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| |
Collapse
|
27
|
Stapornwongkul KS, Vincent JP. Generation of extracellular morphogen gradients: the case for diffusion. Nat Rev Genet 2021; 22:393-411. [PMID: 33767424 DOI: 10.1038/s41576-021-00342-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 02/07/2023]
Abstract
Cells within developing tissues rely on morphogens to assess positional information. Passive diffusion is the most parsimonious transport model for long-range morphogen gradient formation but does not, on its own, readily explain scaling, robustness and planar transport. Here, we argue that diffusion is sufficient to ensure robust morphogen gradient formation in a variety of tissues if the interactions between morphogens and their extracellular binders are considered. A current challenge is to assess how the affinity for extracellular binders, as well as other biophysical and cell biological parameters, determines gradient dynamics and shape in a diffusion-based transport system. Technological advances in genome editing, tissue engineering, live imaging and in vivo biophysics are now facilitating measurement of these parameters, paving the way for mathematical modelling and a quantitative understanding of morphogen gradient formation and modulation.
Collapse
|
28
|
Keil S, Gupta M, Brand M, Knopf F. Heparan sulfate proteoglycan expression in the regenerating zebrafish fin. Dev Dyn 2021; 250:1368-1380. [PMID: 33638212 DOI: 10.1002/dvdy.321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/16/2021] [Accepted: 02/10/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Heparan sulfate proteoglycan (HSPG) expression is found in many animal tissues and regulates growth factor signaling such as of Fibroblast growth factors (Fgf), Wingless/Int (Wnt) and Hedgehog (HH). Glypicans, which are GPI (glycosylphosphatidylinositol)-anchored proteins, and transmembrane-anchored syndecans represent two major HSPG protein families whose involvement in development and disease has been demonstrated. Their participation in regenerative processes both of the central nervous system and of regenerating limbs is well documented. However, whether HSPG are expressed in regenerating zebrafish fins, is currently unknown. RESULTS Here, we carried out a systematic screen of glypican and syndecan mRNA expression in regenerating zebrafish fins during the outgrowth phase. We find that 8 of the 10 zebrafish glypicans and the three known zebrafish syndecans show specific expression at 3 days post amputation. Expression is found in different domains of the regenerate, including the distal and lateral basal layers of the wound epidermis, the distal most blastema and more proximal blastema regions. CONCLUSIONS HSPG expression is prevalent in regenerating zebrafish fins. Further research is needed to delineate the function of glypican and syndecan action during zebrafish fin regeneration.
Collapse
Affiliation(s)
- Sebastian Keil
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany.,Technische Universität Dresden, Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Mansi Gupta
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany.,Merus N.V, Utrecht, Netherlands
| | - Michael Brand
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Franziska Knopf
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany.,Technische Universität Dresden, Center for Healthy Aging TU Dresden, Dresden, Germany
| |
Collapse
|
29
|
Zecca M, Struhl G. A unified mechanism for the control of Drosophila wing growth by the morphogens Decapentaplegic and Wingless. PLoS Biol 2021; 19:e3001111. [PMID: 33657096 PMCID: PMC8148325 DOI: 10.1371/journal.pbio.3001111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 05/25/2021] [Accepted: 01/22/2021] [Indexed: 12/31/2022] Open
Abstract
Development of the Drosophila wing-a paradigm of organ development-is governed by 2 morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Both proteins are produced by defined subpopulations of cells and spread outwards, forming gradients that control gene expression and cell pattern as a function of concentration. They also control growth, but how is unknown. Most studies have focused on Dpp and yielded disparate models in which cells throughout the wing grow at similar rates in response to the grade or temporal change in Dpp concentration or to the different amounts of Dpp "equalized" by molecular or mechanical feedbacks. In contrast, a model for Wg posits that growth is governed by a progressive expansion in morphogen range, via a mechanism in which a minimum threshold of Wg sustains the growth of cells within the wing and recruits surrounding "pre-wing" cells to grow and enter the wing. This mechanism depends on the capacity of Wg to fuel the autoregulation of vestigial (vg)-the selector gene that specifies the wing state-both to sustain vg expression in wing cells and by a feed-forward (FF) circuit of Fat (Ft)/Dachsous (Ds) protocadherin signaling to induce vg expression in neighboring pre-wing cells. Here, we have subjected Dpp to the same experimental tests used to elucidate the Wg model and find that it behaves indistinguishably. Hence, we posit that both morphogens act together, via a common mechanism, to control wing growth as a function of morphogen range.
Collapse
Affiliation(s)
- Myriam Zecca
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| | - Gary Struhl
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| |
Collapse
|
30
|
Scaling a Dpp Morphogen Gradient through Feedback Control of Receptors and Co-receptors. Dev Cell 2021; 53:724-739.e14. [PMID: 32574592 DOI: 10.1016/j.devcel.2020.05.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/23/2020] [Accepted: 05/26/2020] [Indexed: 11/21/2022]
Abstract
Gradients of decapentaplegic (Dpp) pattern Drosophila wing imaginal discs, establishing gene expression boundaries at specific locations. As discs grow, Dpp gradients expand, keeping relative boundary positions approximately stationary. Such scaling fails in mutants for Pentagone (pent), a gene repressed by Dpp that encodes a diffusible protein that expands Dpp gradients. Although these properties fit a recent mathematical model of automatic gradient scaling, that model requires an expander that spreads with minimal loss throughout a morphogen field. Here, we show that Pent's actions are confined to within just a few cell diameters of its site of synthesis and can be phenocopied by manipulating non-diffusible Pent targets strictly within the Pent expression domain. Using genetics and mathematical modeling, we develop an alternative model of scaling driven by feedback downregulation of Dpp receptors and co-receptors. Among the model's predictions is a size beyond which scaling fails-something we observe directly in wing discs.
Collapse
|
31
|
Matusek T, Marcetteau J, Thérond PP. Functions of Wnt and Hedgehog-containing extracellular vesicles in development and disease. J Cell Sci 2020; 133:133/18/jcs209742. [PMID: 32989011 DOI: 10.1242/jcs.209742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Secreted morphogens play a major role in the intercellular communication necessary for animal development. It was initially thought that, in order to organize tissue morphogenesis and control cell fate and proliferation, morphogens diffused freely in the extracellular space. This view has since changed following the discovery that morphogens of the Wnt and Hedgehog (Hh) families are modified by various lipid adducts during their biosynthesis, providing them with high affinity for the membrane bilayer. Recent work performed in model organisms suggests that Wnt and Hh proteins are carried on extracellular vesicles. In this Review, we provide our perspectives on the mechanisms of formation of Wnt- and Hh-containing extracellular vesicles, and discuss their functions during animal development, as well as in various human physiopathologies.
Collapse
Affiliation(s)
- Tamás Matusek
- Université Côte d'Azur, CNRS, INSERM, Institut de Biologie Valrose (iBV), Parc Valrose, 06108 Nice Cedex 2, France
| | - Julien Marcetteau
- Université Côte d'Azur, CNRS, INSERM, Institut de Biologie Valrose (iBV), Parc Valrose, 06108 Nice Cedex 2, France
| | - Pascal P Thérond
- Université Côte d'Azur, CNRS, INSERM, Institut de Biologie Valrose (iBV), Parc Valrose, 06108 Nice Cedex 2, France
| |
Collapse
|
32
|
Linnemannstöns K, Witte L, Karuna M P, Kittel JC, Danieli A, Müller D, Nitsch L, Honemann-Capito M, Grawe F, Wodarz A, Gross JC. Ykt6-dependent endosomal recycling is required for Wnt secretion in the Drosophila wing epithelium. Development 2020; 147:dev.185421. [PMID: 32611603 PMCID: PMC7438013 DOI: 10.1242/dev.185421] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 06/08/2020] [Indexed: 01/09/2023]
Abstract
Morphogens are important signalling molecules for tissue development and their secretion requires tight regulation. In the wing imaginal disc of flies, the morphogen Wnt/Wingless is apically presented by the secreting cell and re-internalized before final long-range secretion. Why Wnt molecules undergo these trafficking steps and the nature of the regulatory control within the endosomal compartment remain unclear. Here, we have investigated how Wnts are sorted at the level of endosomes by the versatile v-SNARE Ykt6. Using in vivo genetics, proximity-dependent proteomics and in vitro biochemical analyses, we show that most Ykt6 is present in the cytosol, but can be recruited to de-acidified compartments and recycle Wnts to the plasma membrane via Rab4-positive recycling endosomes. Thus, we propose a molecular mechanism by which producing cells integrate and leverage endocytosis and recycling via Ykt6 to coordinate extracellular Wnt levels.
Collapse
Affiliation(s)
- Karen Linnemannstöns
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany.,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| | - Leonie Witte
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany.,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| | - Pradhipa Karuna M
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany.,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| | - Jeanette Clarissa Kittel
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany.,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| | - Adi Danieli
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany.,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| | - Denise Müller
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany.,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| | - Lena Nitsch
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany.,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| | - Mona Honemann-Capito
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany.,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| | - Ferdinand Grawe
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Cologne 50931, Germany.,Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases (CECAD), Cologne 50931, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Andreas Wodarz
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Cologne 50931, Germany.,Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases (CECAD), Cologne 50931, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Julia Christina Gross
- Hematology and Oncology, University Medical Centre Goettingen, Goettingen 37075, Germany .,Developmental Biochemistry, University Medical Centre Goettingen, Goettingen 37077, Germany
| |
Collapse
|
33
|
Witte L, Linnemannstöns K, Schmidt K, Honemann-Capito M, Grawe F, Wodarz A, Gross JC. The kinesin motor Klp98A mediates apical to basal Wg transport. Development 2020; 147:dev.186833. [PMID: 32665246 PMCID: PMC7438014 DOI: 10.1242/dev.186833] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 07/02/2020] [Indexed: 12/23/2022]
Abstract
Development and tissue homeostasis rely on the tight regulation of morphogen secretion. In the Drosophila wing imaginal disc epithelium, Wg secretion for long-range signal transduction occurs after apical Wg entry into the endosomal system, followed by secretory endosomal transport. Although Wg release appears to occur from the apical and basal cell sides, its exact post-endocytic fate and the functional relevance of polarized endosomal Wg trafficking are poorly understood. Here, we identify the kinesin-3 family member Klp98A as the master regulator of intracellular Wg transport after apical endocytosis. In the absence of Klp98A, functional mature endosomes accumulate in the apical cytosol, and endosome transport to the basal cytosol is perturbed. Despite the resulting Wg mislocalization, Wg signal transduction occurs normally. We conclude that transcytosis-independent routes for Wg trafficking exist and demonstrate that Wg can be recycled apically via Rab4-recycling endosomes in the absence of Klp98A. Summary: In the polarized wing disc epithelium of Drosophila, Kinesin-like protein 98A mediates transcytosis of multivesicular endosomes.
Collapse
Affiliation(s)
- Leonie Witte
- Hematology and Oncology, University Medical Center Goettingen, 37075 Goettingen, Germany.,Developmental Biochemistry, University Medical Center Goettingen, 37077 Goettingen, Germany
| | - Karen Linnemannstöns
- Hematology and Oncology, University Medical Center Goettingen, 37075 Goettingen, Germany.,Developmental Biochemistry, University Medical Center Goettingen, 37077 Goettingen, Germany
| | - Kevin Schmidt
- Hematology and Oncology, University Medical Center Goettingen, 37075 Goettingen, Germany.,Developmental Biochemistry, University Medical Center Goettingen, 37077 Goettingen, Germany
| | - Mona Honemann-Capito
- Hematology and Oncology, University Medical Center Goettingen, 37075 Goettingen, Germany.,Developmental Biochemistry, University Medical Center Goettingen, 37077 Goettingen, Germany
| | - Ferdinand Grawe
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, 50931 Cologne, Germany.,Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Andreas Wodarz
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, 50931 Cologne, Germany.,Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Julia Christina Gross
- Hematology and Oncology, University Medical Center Goettingen, 37075 Goettingen, Germany .,Developmental Biochemistry, University Medical Center Goettingen, 37077 Goettingen, Germany
| |
Collapse
|
34
|
Orlandi C, Omori Y, Wang Y, Cao Y, Ueno A, Roux MJ, Condomitti G, de Wit J, Kanagawa M, Furukawa T, Martemyanov KA. Transsynaptic Binding of Orphan Receptor GPR179 to Dystroglycan-Pikachurin Complex Is Essential for the Synaptic Organization of Photoreceptors. Cell Rep 2020; 25:130-145.e5. [PMID: 30282023 PMCID: PMC6203450 DOI: 10.1016/j.celrep.2018.08.068] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 08/16/2018] [Accepted: 08/23/2018] [Indexed: 01/05/2023] Open
Abstract
Establishing synaptic contacts between neurons is paramount for nervous system function. This process involves transsynaptic interactions between a host of cell adhesion molecules that act in cooperation with the proteins of the extracellular matrix to specify uniquephysiological propertiesofindividual synaptic connections. However, understanding of the molecular mechanisms that generate functional diversity in an input-specific fashion is limited. In this study, we identify that major components of the extracellular matrix proteins present in the synaptic cleft—members oftheheparansulfateproteoglycan (HSPG) family—associate with the GPR158/179 group of orphan receptors. Using the mammalian retina as a model system, we demonstrate that the HSPG member Pikachurin, released by photoreceptors, recruits a key post-synaptic signaling complex of downstream ON-bipolar neurons in coordination with the presynaptic dystroglycan glycoprotein complex. We further demonstrate that this transsynaptic assembly plays an essential role in synaptic transmission of photoreceptor signals. Orlandi et al. identify transsynaptic assembly at photoreceptor synapses involving pre-synaptic dystrophindystroglycan complex and the postsynaptic orphan receptor GPR179 bridged by HSPG protein Pikachurin in the cleft and demonstrate its role in shaping transmission of photoreceptor signals.
Collapse
Affiliation(s)
- Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Yoshihiro Omori
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Yuchen Wang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Akiko Ueno
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Michel J Roux
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Centre National de la Recherche Scientifique, UMR7104, INSERM, U1258, Illkirch, France
| | - Giuseppe Condomitti
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Herestraat 49, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Herestraat 49, 3000 Leuven, Belgium
| | - Motoi Kanagawa
- Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
35
|
Characterization of C. elegans Chondroitin Proteoglycans and Their Large Functional and Structural Heterogeneity; Evolutionary Aspects on Structural Differences Between Humans and the Nematode. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 21:155-170. [PMID: 32185697 DOI: 10.1007/5584_2020_485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Proteoglycans regulate important cellular pathways in essentially all metazoan organisms. While considerable effort has been devoted to study structural and functional aspects of proteoglycans in vertebrates, the knowledge of the core proteins and proteoglycan-related functions in invertebrates is relatively scarce, even for C.elegans. This nematode produces a large amount of non-sulfated chondroitin in addition to small amount of low-sulfated chondroitin chains (Chn and CS chains, respectively). Until recently, 9 chondroitin core proteins (CPGs) had been identified in C.elegans, none of which showed any homology to vertebrate counterparts or to other invertebrate core proteins. By using a glycoproteomic approach, we recently characterized the chondroitin glycoproteome of C.elegans, resulting in the identification of 15 novel CPG core proteins in addition to the 9 previously established. Three of the novel core proteins displayed homology to human proteins, indicating that CPG and CSPG core proteins may be more conserved throughout evolution than previously perceived. Bioinformatic analysis of the primary amino acid sequences revealed that the core proteins contained a broad range of functional domains, indicating that specialization of proteoglycan-mediated functions may have evolved early in metazoan evolution. This review specifically discusses our recent data in relation to previous knowledge of core proteins and GAG-attachment sites in Chn and CS proteoglycans of C.elegans and humans, and point out both converging and diverging aspects of proteoglycan evolution.
Collapse
|
36
|
Won JH, Kim GW, Kim JY, Cho DG, Kwon B, Bae YK, Cho KO. ADAMTS Sol narae cleaves extracellular Wingless to generate a novel active form that regulates cell proliferation in Drosophila. Cell Death Dis 2019; 10:564. [PMID: 31332194 PMCID: PMC6646336 DOI: 10.1038/s41419-019-1794-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 06/14/2019] [Accepted: 07/04/2019] [Indexed: 11/23/2022]
Abstract
Wnt/ Wingless (Wg) is essential for embryonic development and adult homeostasis in all metazoans, but the mechanisms by which secreted Wnt/Wg is processed remain largely unknown. A Drosophila Sol narae (Sona) is a member of ADisintegrin And Metalloprotease with ThromboSpondin motif (ADAMTS) family, and positively regulates Wg signaling by promoting Wg secretion. Here we report that Sona and Wg are secreted by both conventional Golgi and exosomal transports, and Sona cleaves extracellular Wg at the two specific sites, leading to the generation of N-terminal domain (NTD) and C-terminal domain (CTD) fragments. The cleaved forms of extracellular Wg were detected in the extracellular region of fly wing discs, and its level was substantially reduced in sona mutants. Transient overexpression of Wg-CTD increased wing size while prolonged overexpression caused lethality and developmental defects. In contrast, Wg-NTD did not induce any phenotype. Moreover, the wing defects and lethality induced by sona RNAi were considerably rescued by Wg-CTD, indicating that a main function of extracellular Sona is the generation of Wg-CTD. Wg-CTD stabilized cytoplasmic Armadillo (Arm) and had genetic interactions with components of canonical Wg signaling. Wg-CTD also induced Wg downstream targets such as Distal-less (Dll) and Vestigial (Vg). Most importantly, Cyclin D (Cyc D) was induced by Wg-CTD but not by full-length Wg. Because Sona also induces Cyc D in a cell non-autonomous manner, Wg-CTD generated by Sona in the extracellular region activates a subset of Wg signaling whose major function is the regulation of cell proliferation.
Collapse
Affiliation(s)
- Jong-Hoon Won
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, Korea
| | - Go-Woon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, Korea
| | - Ja-Young Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, Korea
| | - Dong-Gyu Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, Korea
| | - Buki Kwon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, Korea
| | - Young-Kyung Bae
- Center for Bioanalysis, Korea Research Institute of Standards and Science, 267 Gajung-ro, Yuseung-gu, Daejeon, Korea
| | - Kyung-Ok Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, Korea.
| |
Collapse
|
37
|
Torres VI, Godoy JA, Inestrosa NC. Modulating Wnt signaling at the root: Porcupine and Wnt acylation. Pharmacol Ther 2019; 198:34-45. [DOI: 10.1016/j.pharmthera.2019.02.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/12/2019] [Indexed: 02/07/2023]
|
38
|
Fernández D, Guereño M, Lago Huvelle MA, Cercato M, Peters MG. Signaling network involved in the GPC3-induced inhibition of breast cancer progression: role of canonical Wnt pathway. J Cancer Res Clin Oncol 2018; 144:2399-2418. [PMID: 30267212 DOI: 10.1007/s00432-018-2751-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/11/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE We have shown that GPC3 overexpression in breast cancer cells inhibits in vivo tumor progression, by acting as a metastatic suppressor. GPC3-overexpressing cells are less clonogenic, viable and motile, while their homotypic adhesion is increased. We have presented evidences indicating that GPC3 inhibits canonical Wnt and Akt pathways, while non-canonical Wnt and p38MAPK cascades are activated. In this study, we aimed to investigate whether GPC3-induced Wnt signaling inhibition modulates breast cancer cell properties as well as to describe the interactions among pathways modulated by GPC3. METHODS Fluorescence microscopy, qRT-PCR microarray, gene reporter assay and Western blotting were performed to determine gene expression levels, signaling pathway activities and molecule localization. Lithium was employed to activate canonical Wnt pathway and treated LM3-GPC3 cell viability, migration, cytoskeleton organization and homotypic adhesion were assessed using MTS, wound healing, phalloidin staining and suspension growth assays, respectively. RESULTS We provide new data demonstrating that GPC3 blocks-also at a transcriptional level-both autocrine and paracrine canonical Wnt activities, and that this inhibition is required for GPC3 to modulate migration and homotypic adhesion. Our results indicate that GPC3 is secreted into the extracellular media, suggesting that secreted GPC3 competes with Wnt factors or interacts with them and thus prevents Wnt binding to Fz receptors. We also describe the complex network of interactions among GPC3-modulated signaling pathways. CONCLUSION GPC3 is operating through an intricate molecular signaling network. From the balance of these interactions, the inhibition of breast metastatic spread induced by GPC3 emerges.
Collapse
Affiliation(s)
- Dolores Fernández
- Cell Biology Department, Research Area, Institute of Oncology "Angel H. Roffo", University of Buenos Aires, Av. San Martín 5481, C1417DTB, Buenos Aires, Argentina
| | - Macarena Guereño
- Cell Biology Department, Research Area, Institute of Oncology "Angel H. Roffo", University of Buenos Aires, Av. San Martín 5481, C1417DTB, Buenos Aires, Argentina
| | - María Amparo Lago Huvelle
- Cell Biology Department, Research Area, Institute of Oncology "Angel H. Roffo", University of Buenos Aires, Av. San Martín 5481, C1417DTB, Buenos Aires, Argentina
| | - Magalí Cercato
- Cell Biology Department, Research Area, Institute of Oncology "Angel H. Roffo", University of Buenos Aires, Av. San Martín 5481, C1417DTB, Buenos Aires, Argentina
| | - María Giselle Peters
- Cell Biology Department, Research Area, Institute of Oncology "Angel H. Roffo", University of Buenos Aires, Av. San Martín 5481, C1417DTB, Buenos Aires, Argentina.
- National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
39
|
Yamamoto-Hino M, Katsumata E, Suzuki E, Maeda Y, Kinoshita T, Goto S. Nuclear envelope localization of PIG-B is essential for GPI-anchor synthesis in Drosophila. J Cell Sci 2018; 131:jcs.218024. [PMID: 30266758 PMCID: PMC6215393 DOI: 10.1242/jcs.218024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 09/21/2018] [Indexed: 12/14/2022] Open
Abstract
Membrane lipid biosynthesis is a complex process that takes place in various intracellular compartments. Glycosylphosphatidylinositol (GPI), a lipid involved in membrane anchoring of some proteins, is synthesized by the PIG enzymes. Most PIGs are localized to the endoplasmic reticulum (ER), but Drosophila PIG-B (DmPIG-B) is localized to the nuclear envelope (NE). To determine whether the NE localization of DmPIG-B is functionally important, we defined the determinants of localization and generated an ER-localized form, denoted DmPIG-B[ER]. The enzymatic activity of DmPIG-B[ER] was comparable to that of NE-localized DmPIG-B[NE]. Expression of DmPIG-B[ER] inefficiently rescued the lethality of the PIG-B mutant, whereas DmPIG-B[NE] rescued this lethality fully. DmPIG-B[ER] was preferentially degraded by lysosomes, suggesting that the NE localization is essential for function and stability of the protein. In addition, we found that the region of the ER proximal to the NE is the site of translation of GPI-anchored proteins and addition of GPI. Thus, the NE and proximal ER may provide a platform for efficient GPI anchoring. Summary: In Drosophila, localization of the enzyme PIG-B in the nuclear envelope (NE) is important for GPI anchor synthesis, and the NE and the perinuclear ER form a platform for the GPI modification.
Collapse
Affiliation(s)
- Miki Yamamoto-Hino
- Department of Life Science, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| | - Eri Katsumata
- Department of Life Science, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| | - Emiko Suzuki
- Gene Network Laboratory, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Yusuke Maeda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Taroh Kinoshita
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Satoshi Goto
- Department of Life Science, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| |
Collapse
|
40
|
Hogvall M, Budd GE, Janssen R. Gene expression analysis of potential morphogen signalling modifying factors in Panarthropoda. EvoDevo 2018; 9:20. [PMID: 30288252 PMCID: PMC6162966 DOI: 10.1186/s13227-018-0109-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/04/2018] [Indexed: 11/28/2022] Open
Abstract
Background Morphogen signalling represents a key mechanism of developmental processes during animal development. Previously, several evolutionary conserved morphogen signalling pathways have been identified, and their players such as the morphogen receptors, morphogen modulating factors (MMFs) and the morphogens themselves have been studied. MMFs are factors that regulate morphogen distribution and activity. The interactions of MMFs with different morphogen signalling pathways such as Wnt signalling, Hedgehog (Hh) signalling and Decapentaplegic (Dpp) signalling are complex because some of the MMFs have been shown to interact with more than one signalling pathway, and depending on genetic context, to have different, biphasic or even opposing function. This complicates the interpretation of expression data and functional data of MMFs and may be one reason why data on MMFs in other arthropods than Drosophila are scarce or totally lacking. Results As a first step to a better understanding of the potential roles of MMFs in arthropod development, we investigate here the embryonic expression patterns of division abnormally delayed (dally), dally-like protein (dlp), shifted (shf) and secreted frizzled-related protein 125 (sFRP125) and sFRP34 in the beetle Tribolium castaneum, the spider Parasteatoda tepidariorum, the millipede Glomeris marginata and the onychophoran Euperipatoides kanangrensis. This pioneer study represents the first comprehensive comparative data set of these genes in panarthropods. Conclusions Expression profiles reveal a high degree of diversity, suggesting that MMFs may represent highly evolvable nodes in otherwise conserved gene regulatory networks. Conserved aspects of MMF expression, however, appear to concern function in segmentation and limb development, two of the key topics of evolutionary developmental research. Electronic supplementary material The online version of this article (10.1186/s13227-018-0109-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mattias Hogvall
- Department of Earth Sciences, Palaeobiology, Uppsala University, Villavägen 16, Uppsala, Sweden
| | - Graham E Budd
- Department of Earth Sciences, Palaeobiology, Uppsala University, Villavägen 16, Uppsala, Sweden
| | - Ralf Janssen
- Department of Earth Sciences, Palaeobiology, Uppsala University, Villavägen 16, Uppsala, Sweden
| |
Collapse
|
41
|
Wingless Signaling: A Genetic Journey from Morphogenesis to Metastasis. Genetics 2018; 208:1311-1336. [PMID: 29618590 DOI: 10.1534/genetics.117.300157] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 12/13/2017] [Indexed: 12/15/2022] Open
Abstract
This FlyBook chapter summarizes the history and the current state of our understanding of the Wingless signaling pathway. Wingless, the fly homolog of the mammalian Wnt oncoproteins, plays a central role in pattern generation during development. Much of what we know about the pathway was learned from genetic and molecular experiments in Drosophila melanogaster, and the core pathway works the same way in vertebrates. Like most growth factor pathways, extracellular Wingless/Wnt binds to a cell surface complex to transduce signal across the plasma membrane, triggering a series of intracellular events that lead to transcriptional changes in the nucleus. Unlike most growth factor pathways, the intracellular events regulate the protein stability of a key effector molecule, in this case Armadillo/β-catenin. A number of mysteries remain about how the "destruction complex" destabilizes β-catenin and how this process is inactivated by the ligand-bound receptor complex, so this review of the field can only serve as a snapshot of the work in progress.
Collapse
|
42
|
Vuillaume ML, Moizard MP, Rossignol S, Cottereau E, Vonwill S, Alessandri JL, Busa T, Colin E, Gérard M, Giuliano F, Lambert L, Lefevre M, Kotecha U, Nampoothiri S, Netchine I, Raynaud M, Brioude F, Toutain A. Mutation update for the GPC3 gene involved in Simpson-Golabi-Behmel syndrome and review of the literature. Hum Mutat 2018; 39:790-805. [PMID: 29637653 DOI: 10.1002/humu.23428] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/22/2018] [Accepted: 04/02/2018] [Indexed: 11/08/2022]
Abstract
Simpson-Golabi-Behmel syndrome (SGBS) is an X-linked multiple congenital anomalies and overgrowth syndrome caused by a defect in the glypican-3 gene (GPC3). Until now, GPC3 mutations have been reported in isolated cases or small series and the global genotypic spectrum of these mutations has never been delineated. In this study, we review the 57 previously described GPC3 mutations and significantly expand this mutational spectrum with the description of 29 novel mutations. Compiling our data and those of the literature, we provide an overview of 86 distinct GPC3 mutations identified in 120 unrelated families, ranging from single nucleotide variations to complex genomic rearrangements and dispersed throughout the entire coding region of GPC3. The vast majority of them are deletions or truncating mutations (frameshift, nonsense mutations) predicted to result in a loss-of-function. Missense mutations are rare and the two which were functionally characterized, impaired GPC3 function by preventing GPC3 cleavage and cell surface addressing respectively. This report by describing for the first time the wide mutational spectrum of GPC3 could help clinicians and geneticists in interpreting GPC3 variants identified incidentally by high-throughput sequencing technologies and also reinforces the need for functional validation of non-truncating mutations (missense, in frame mutations, duplications).
Collapse
Affiliation(s)
- Marie-Laure Vuillaume
- Service de Génétique, CHU de Tours, Hôpital Bretonneau, Tours, France.,INSERM UMR_U930, Faculté de Médecine, Université de Tours, Tours, France
| | - Marie-Pierre Moizard
- Service de Génétique, CHU de Tours, Hôpital Bretonneau, Tours, France.,INSERM UMR_U930, Faculté de Médecine, Université de Tours, Tours, France
| | - Sylvie Rossignol
- Unité d'explorations fonctionnelles endocriniennes, CHU Paris Est, Hôpital d'Enfants Armand-Trousseau, Paris, France.,Service de génétique médicale, CHU de Strasbourg, Hôpital de Hautepierre, Strasbourg, France
| | - Edouard Cottereau
- Service de Génétique, CHU de Tours, Hôpital Bretonneau, Tours, France
| | - Sandrine Vonwill
- Service de Génétique, CHU de Tours, Hôpital Bretonneau, Tours, France.,INSERM UMR_U930, Faculté de Médecine, Université de Tours, Tours, France
| | | | - Tiffany Busa
- Unité de Génétique Clinique, Département de génétique médicale, Hôpital de la Timone, CHU de Marseille, Marseille, France
| | - Estelle Colin
- Département de biochimie et génétique, CHU d'Angers, Angers, France
| | - Marion Gérard
- Service de génétique, CHU de Caen, Hôpital Clémenceau, Avenue Georges Clémenceau, Caen, France
| | - Fabienne Giuliano
- Service de génétique médicale, CHU de Nice, Hôpital l'Archet 2, Nice, France
| | - Laetitia Lambert
- Service de Génétique Clinique, Hôpital d'Enfants, CHU de Nancy, Rue du Morvan, Vandoeuvre-Lès-Nancy, France
| | - Mathilde Lefevre
- Centre de génétique, Hôpital d'enfants, CHU Dijon Bourgogne, Dijon, France
| | - Udhaya Kotecha
- Center of Medical Genetics, Sir Ganga Ram Hospital, Rajinder Nagar, New Delhi, India
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences and Research Center, AIMS Poneakara P O, Cochin, Kerala, India
| | - Irène Netchine
- Unité d'explorations fonctionnelles endocriniennes, CHU Paris Est, Hôpital d'Enfants Armand-Trousseau, Paris, France
| | - Martine Raynaud
- Service de Génétique, CHU de Tours, Hôpital Bretonneau, Tours, France.,INSERM UMR_U930, Faculté de Médecine, Université de Tours, Tours, France
| | - Frédéric Brioude
- Unité d'explorations fonctionnelles endocriniennes, CHU Paris Est, Hôpital d'Enfants Armand-Trousseau, Paris, France
| | - Annick Toutain
- Service de Génétique, CHU de Tours, Hôpital Bretonneau, Tours, France.,INSERM UMR_U930, Faculté de Médecine, Université de Tours, Tours, France
| |
Collapse
|
43
|
Syndecan-1 promotes Wnt/β-catenin signaling in multiple myeloma by presenting Wnts and R-spondins. Blood 2018; 131:982-994. [DOI: 10.1182/blood-2017-07-797050] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/01/2017] [Indexed: 02/07/2023] Open
Abstract
Key Points
HS chains decorating syndecan-1 promote autocrine and paracrine Wnt signaling in MM. Loss of HS inhibits MM cell growth by attenuating Wnt signaling.
Collapse
|
44
|
Neuronal-specific impairment of heparan sulfate degradation in Drosophila reveals pathogenic mechanisms for Mucopolysaccharidosis type IIIA. Exp Neurol 2018; 303:38-47. [PMID: 29408731 DOI: 10.1016/j.expneurol.2018.01.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/13/2017] [Accepted: 01/31/2018] [Indexed: 01/17/2023]
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is a lysosomal storage disorder resulting from the deficit of the N-sulfoglucosamine sulfohydrolase (SGSH) enzyme that leads to accumulation of partially-degraded heparan sulfate. MPS IIIA is characterized by severe neurological symptoms, clinically presenting as Sanfilippo syndrome, for which no effective therapy is available. The lysosomal SGSH enzyme is conserved in Drosophila and we have identified increased levels of heparan sulfate in flies with ubiquitous knockdown of SGSH/CG14291. Using neuronal specific knockdown of SGSH/CG14291 we have also observed a higher abundance of Lysotracker-positive puncta as well as increased expression of GFP tagged Ref(2)P supporting disruption to lysosomal function. We have also observed a progressive defect in climbing ability, a hallmark of neurological dysfunction. Genetic screens indicate proteins and pathways that can functionally modify the climbing phenotype, including autophagy-related proteins (Atg1 and Atg18), superoxide dismutase enzymes (Sod1 and Sod2) and heat shock protein (HSPA1). In addition, reducing heparan sulfate biosynthesis by knocking down sulfateless or slalom expression significantly worsens the phenotype; an important observation given that substrate inhibition is being evaluated clinically as a treatment for MPS IIIA. Identifying the cellular pathways that can modify MPS IIIA neuropathology is an essential step in the development of novel therapeutic approaches to prevent and/or ameliorate symptoms in children with Sanfilippo syndrome.
Collapse
|
45
|
Dear ML, Shilts J, Broadie K. Neuronal activity drives FMRP- and HSPG-dependent matrix metalloproteinase function required for rapid synaptogenesis. Sci Signal 2017; 10:eaan3181. [PMID: 29114039 PMCID: PMC5743058 DOI: 10.1126/scisignal.aan3181] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Matrix metalloproteinase (MMP) functions modulate synapse formation and activity-dependent plasticity. Aberrant MMP activity is implicated in fragile X syndrome (FXS), a disease caused by the loss of the RNA-binding protein FMRP and characterized by neurological dysfunction and intellectual disability. Gene expression studies in Drosophila suggest that Mmps cooperate with the heparan sulfate proteoglycan (HSPG) glypican co-receptor Dally-like protein (Dlp) to restrict trans-synaptic Wnt signaling and that synaptogenic defects in the fly model of FXS are alleviated by either inhibition of Mmp or genetic reduction of Dlp. We used the Drosophila neuromuscular junction (NMJ) glutamatergic synapse to test activity-dependent Dlp and Mmp intersections in the context of FXS. We found that rapid, activity-dependent synaptic bouton formation depended on secreted Mmp1. Acute neuronal stimulation reduced the abundance of Mmp2 but increased that of both Mmp1 and Dlp, as well as enhanced the colocalization of Dlp and Mmp1 at the synapse. Dlp function promoted Mmp1 abundance, localization, and proteolytic activity around synapses. Dlp glycosaminoglycan (GAG) chains mediated this functional interaction with Mmp1. In the FXS fly model, activity-dependent increases in Mmp1 abundance and activity were lost but were restored by reducing the amount of synaptic Dlp. The data suggest that neuronal activity-induced, HSPG-dependent Mmp regulation drives activity-dependent synaptogenesis and that this is impaired in FXS. Thus, exploring this mechanism further may reveal therapeutic targets that have the potential to restore synaptogenesis in FXS patients.
Collapse
Affiliation(s)
- Mary L Dear
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Jarrod Shilts
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232, USA.
- Department of Cell and Developmental Biology, Vanderbilt University and Medical School, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University and Medical School, Nashville, TN 37232, USA
| |
Collapse
|
46
|
Reynolds-Peterson CE, Zhao N, Xu J, Serman TM, Xu J, Selleck SB. Heparan sulfate proteoglycans regulate autophagy in Drosophila. Autophagy 2017; 13:1262-1279. [PMID: 28402693 PMCID: PMC5584867 DOI: 10.1080/15548627.2017.1304867] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 02/22/2017] [Accepted: 03/06/2017] [Indexed: 12/12/2022] Open
Abstract
Heparan sulfate-modified proteoglycans (HSPGs) are important regulators of signaling and molecular recognition at the cell surface and in the extracellular space. Disruption of HSPG core proteins, HS-synthesis, or HS-degradation can have profound effects on growth, patterning, and cell survival. The Drosophila neuromuscular junction provides a tractable model for understanding the activities of HSPGs at a synapse that displays developmental and activity-dependent plasticity. Muscle cell-specific knockdown of HS biosynthesis disrupted the organization of a specialized postsynaptic membrane, the subsynaptic reticulum (SSR), and affected the number and morphology of mitochondria. We provide evidence that these changes result from a dysregulation of macroautophagy (hereafter referred to as autophagy). Cellular and molecular markers of autophagy are all consistent with an increase in the levels of autophagy in the absence of normal HS-chain biosynthesis and modification. HS production is also required for normal levels of autophagy in the fat body, the central energy storage and nutritional sensing organ in Drosophila. Genetic mosaic analysis indicates that HS-dependent regulation of autophagy occurs non-cell autonomously, consistent with HSPGs influencing this cellular process via signaling in the extracellular space. These findings demonstrate that HS biosynthesis has important regulatory effects on autophagy and that autophagy is critical for normal assembly of postsynaptic membrane specializations.
Collapse
Affiliation(s)
- Claire E. Reynolds-Peterson
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Na Zhao
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Jie Xu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Taryn M. Serman
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Jielin Xu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Scott B. Selleck
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
47
|
Saied-Santiago K, Townley RA, Attonito JD, da Cunha DS, Díaz-Balzac CA, Tecle E, Bülow HE. Coordination of Heparan Sulfate Proteoglycans with Wnt Signaling To Control Cellular Migrations and Positioning in Caenorhabditis elegans. Genetics 2017; 206:1951-1967. [PMID: 28576860 PMCID: PMC5560800 DOI: 10.1534/genetics.116.198739] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 05/20/2017] [Indexed: 11/18/2022] Open
Abstract
Heparan sulfates (HS) are linear polysaccharides with complex modification patterns, which are covalently bound via conserved attachment sites to core proteins to form heparan sulfate proteoglycans (HSPGs). HSPGs regulate many aspects of the development and function of the nervous system, including cell migration, morphology, and network connectivity. HSPGs function as cofactors for multiple signaling pathways, including the Wnt-signaling molecules and their Frizzled receptors. To investigate the functional interactions among the HSPG and Wnt networks, we conducted genetic analyses of each, and also between these networks using five cellular migrations in the nematode Caenorhabditis elegans We find that HSPG core proteins act genetically in a combinatorial fashion dependent on the cellular contexts. Double mutant analyses reveal distinct redundancies among HSPGs for different migration events, and different cellular migrations require distinct heparan sulfate modification patterns. Our studies reveal that the transmembrane HSPG SDN-1/Syndecan functions within the migrating cell to promote cellular migrations, while the GPI-linked LON-2/Glypican functions cell nonautonomously to establish the final cellular position. Genetic analyses with the Wnt-signaling system show that (1) a given HSPG can act with different Wnts and Frizzled receptors, and that (2) a given Wnt/Frizzled pair acts with different HSPGs in a context-dependent manner. Lastly, we find that distinct HSPG and Wnt/Frizzled combinations serve separate functions to promote cellular migration and establish position of specific neurons. Our studies suggest that HSPGs use structurally diverse glycans in coordination with Wnt-signaling pathways to control multiple cellular behaviors, including cellular and axonal migrations and, cellular positioning.
Collapse
Affiliation(s)
| | - Robert A Townley
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - John D Attonito
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Dayse S da Cunha
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Carlos A Díaz-Balzac
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Eillen Tecle
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
48
|
Xia W, Hu J, Liu F, Ma J, Sun S, Zhang J, Jin K, Huang J, Jiang N, Wang X, Li W, Ma Z, Ma D. New role of LRP5, associated with nonsyndromic autosomal-recessive hereditary hearing loss. Hum Mutat 2017; 38:1421-1431. [PMID: 28677207 DOI: 10.1002/humu.23285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 06/19/2017] [Accepted: 06/25/2017] [Indexed: 12/14/2022]
Abstract
Human hearing loss is a common neurosensory disorder about which many basic research and clinically relevant questions are unresolved. At least 50% of hearing loss are due to a genetic etiology. Although hundreds of genes have been reported, there are still hundreds of related deafness genes to be found. Clinical, genetic, and functional investigations were performed to identify the causative mutation in a distinctive Chinese family with postlingual nonsyndromic sensorineural hearing loss. Whole-exome sequencing (WES) identified lipoprotein receptor-related protein 5 (LRP5), a member of the low-density lipoprotein receptor family, as the causative gene in this family. In the zebrafish model, lrp5 downregulation using morpholinos led to significant abnormalities in zebrafish inner ear and lateral line neuromasts and contributed, to some extent, to disabilities in hearing and balance. Rescue experiments showed that LRP5 mutation is associated with hearing loss. Knocking down lrp5 in zebrafish results in reduced expression of several genes linked to Wnt signaling pathway and decreased cell proliferation when compared with those in wild-type zebrafish. In conclusion, the LRP5 mutation influences cell proliferation through the Wnt signaling pathway, thereby reducing the number of supporting cells and hair cells and leading to nonsyndromic hearing loss in this Chinese family.
Collapse
Affiliation(s)
- Wenjun Xia
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Jiongjiong Hu
- Department of Otorhinolaryngology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Fei Liu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jing Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Center Laboratory, Bao'an Maternal and Children Healthcare Hospital, Key Laboratory of Birth Defects Research, Shenzhen, China
| | - Shaoyang Sun
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kaiyue Jin
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianbo Huang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Nan Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xu Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wen Li
- Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Zhaoxin Ma
- Department of Otorhinolaryngology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Duan Ma
- Institutes of Biomedical Science, Fudan University, Shanghai, China.,Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, Collaborative Innovation Center of Genetics and Development, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Children's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Pazina T, Shemesh A, Brusilovsky M, Porgador A, Campbell KS. Regulation of the Functions of Natural Cytotoxicity Receptors by Interactions with Diverse Ligands and Alterations in Splice Variant Expression. Front Immunol 2017; 8:369. [PMID: 28424697 PMCID: PMC5371597 DOI: 10.3389/fimmu.2017.00369] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/14/2017] [Indexed: 12/11/2022] Open
Abstract
The natural cytotoxicity receptor (NCR) family is constituted by NKp46, NKp44, and NKp30 in humans, which are expressed mainly on natural killer (NK) cells and are encoded by the ncr1, ncr2, and ncr3 genes, respectively. NCRs have classically been defined as activating receptors that trigger cytotoxicity and cytokine responses by NK cells upon engaging with ligands on tumor cells. Several new findings, however, have challenged this model and identified alternative mechanisms regulating the function of NCRs. Recent reports indicate that ligand matters, since the interaction of NKp44 with distinct ligands on target cells can either activate or inhibit NK cells. Also, the NCRs have been found to interact with distinct specificities to various heparan sulfate glycosaminoglycans, which are complex polysaccharides found in extracellular matrix or on cell surface heparan sulfate proteoglycans (HSPGs). The NCRs can engage with HSPGs in trans as a co-ligand on the target cells or in cis on the NK cell surface to regulate receptor–ligand interactions and NK cell activation. A number of splice variants of ncr2 and ncr3 have also been identified, and a predominant expression of certain variants results in inhibitory signaling through NKp44 and NKp30. Several recent studies have found that the selective expression of some of these inhibitory splice variants can significantly influence outcome in the contexts of cancer, infection, and pregnancy. These findings establish that NCR functions are more diverse than originally thought, and better understanding of their splice variant expression profiles and ligand interactions are needed to establish their functional regulation in the context of human health.
Collapse
Affiliation(s)
- Tatiana Pazina
- Blood Cell Development and Function Program, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA.,Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", St. Petersburg, Russia
| | - Avishai Shemesh
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michael Brusilovsky
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Angel Porgador
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kerry S Campbell
- Blood Cell Development and Function Program, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
50
|
Yukita A, Hara M, Hosoya A, Nakamura H. Relationship between localization of proteoglycans and induction of neurotrophic factors in mouse dental pulp. J Oral Biosci 2017. [DOI: 10.1016/j.job.2016.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|