1
|
Wu J, Fu J, Li X, Xiong F, Yang F. Down-regulation of MYO1A inhibits trophoblast cell proliferation and migration through SMURF2/Hedgehog signaling pathway and leads to fetal growth restriction. Placenta 2025; 164:73-83. [PMID: 40147359 DOI: 10.1016/j.placenta.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
INTRODUCTION Fetal growth restriction (FGR) is most commonly related to insufficient placental perfusion caused by insufficient trophoblast proliferation and migration. Myosin Ia, encoded by the gene MYO1A, plays an important role in cytoskeleton recombination and cell movement. In this study, we found that downregulation of MYO1A inhibits Hedgehog (Hh) signaling by interacting with SMURF2 in choriocarcinoma cells, leading to FGR. METHODS A total of 59 placenta samples (26 FGR placentas and 33 normal placentas) were collected. The expression of MYO1A in placental tissues of the two groups was detected by qRT-PCR and Western blotting. The proliferation ability of choriocarcinoma cell lines HTR-8/SVneo and JEG3 was tested by CCK8 and colony formation experiments, and the migration ability was tested by transwell and wound healing experiments. Co-immunoprecipitation assay is used to verify the interaction between myosin Ia and SMURF2. RESULT We found that MYO1A expression was significantly lower in the placentas of pregnant women with FGR than in normal pregnant women. Moreover, the knockdown of MYO1A has been observed to inhibit choriocarcinoma cells proliferation and migration. Downregulation of MYO1A inhibits Hh signaling by reducing SMURF2 expression. DISCUSSION Our findings suggest that FGR is associated with a down-regulation of MYO1A, which may affect the Hh pathway through its interaction with SMURF2. This provides clues for a deeper understanding of the specific mechanisms underlying FGR.
Collapse
Affiliation(s)
- Jie Wu
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiahui Fu
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyun Li
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fu Xiong
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Fang Yang
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
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
|
3
|
Zhang J, Liu Y, Wang C, Vander Kooi CW, Jia J. Phosphatidic acid binding to Patched contributes to the inhibition of Smoothened and Hedgehog signaling in Drosophila wing development. Sci Signal 2023; 16:eadd6834. [PMID: 37847757 PMCID: PMC10661859 DOI: 10.1126/scisignal.add6834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/25/2023] [Indexed: 10/19/2023]
Abstract
Hedgehog (Hh) signaling controls growth and patterning during embryonic development and homeostasis in adult tissues. Hh binding to the receptor Patched (Ptc) elicits intracellular signaling by relieving Ptc-mediated inhibition of the transmembrane protein Smoothened (Smo). We uncovered a role for the lipid phosphatidic acid (PA) in the regulation of the Hh pathway in Drosophila melanogaster. Deleting the Ptc C-terminal tail or mutating the predicted PA-binding sites within it prevented Ptc from inhibiting Smo in wing discs and in cultured cells. The C-terminal tail of Ptc directly interacted with PA in vitro, an association that was reduced by Hh, and increased the amount of PA at the plasma membrane in cultured cells. Smo also interacted with PA in vitro through a binding pocket located in the transmembrane region, and mutating residues in this pocket reduced Smo activity in vivo and in cells. By genetically manipulating PA amounts in vivo or treating cultured cells with PA, we demonstrated that PA promoted Smo activation. Our findings suggest that Ptc may sequester PA in the absence of Hh and release it in the presence of Hh, thereby increasing the amount of PA that is locally available to promote Smo activation.
Collapse
Affiliation(s)
- Jie Zhang
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Yajuan Liu
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Craig W. Vander Kooi
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jianhang Jia
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| |
Collapse
|
4
|
Gao Y, Shan Z, Jian C, Wang Y, Yao X, Li S, Ti X, Zhao G, Liu C, Zhang Q. HIB/SPOP inhibits Ci/Gli-mediated tumorigenesis by modulating the RNA Polymerase II components stabilities. iScience 2023; 26:107334. [PMID: 37554435 PMCID: PMC10404538 DOI: 10.1016/j.isci.2023.107334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/09/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Hedgehog (Hh) signaling mediated by transcription factor Ci/Gli plays a vital role in embryonic development and adult tissue homeostasis in invertebrates and vertebrates, whose dysregulation leads to many human disorders, including cancer. However, till now, cofactors of Ci/Gli which can affect tumorigenesis are not well known. Here, through genetic screen, we find overexpression of active Ci alone is not sufficient to generate tumor-like eye phenotype in Drosophila, however, its overexpression combined with knockdown of hib causes a striking tumor-like big eye phenotype. Mechanistically, HIB/SPOP inhibits Ci/Gli-mediated tumorigenesis by modulating the RNA polymerase II (RNAPII) components Rpb3/Rpb7 stabilities in E3 ligase dependent manner. In addition, Ci/Gli can promote HIB/SPOP-mediated Rpb7/Rpb3 degradation. Taken together, our results indicate Ci/Gli needs to hook up with suitable RNAPII together to achieve the tumor-like eye phenotype and HIB/SPOP plays dual roles through controlling Ci/Gli and Rpb3/Rpb7 protein stabilities to temper Ci/Gli/RNAPII-mediated tumorigenesis.
Collapse
Affiliation(s)
- Yuxue Gao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Zhaoliang Shan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Chunhua Jian
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Ying Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Xia Yao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Shengnan Li
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Xiuxiu Ti
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Guochun Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| | - Chen Liu
- Department of Medical Genetics, Nanjing Medical University, Nanjing 211166, China
| | - Qing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
| |
Collapse
|
5
|
Jiang Z, Qu L, Cui G, Zhong G. Smoothened antagonist sonidegib affects the development of D. melanogaster larvae via suppression of epidermis formation. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 194:105462. [PMID: 37532307 DOI: 10.1016/j.pestbp.2023.105462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 08/04/2023]
Abstract
Hedgehog (Hh) signaling is essential for the regulation of embryonic growth and development, the maintenance of stem cell autostasis, and tissue formation, whether in vertebrates or invertebrates. However, exploration into the Hh pathway antagonists in Drosophila or other pests of agricultural importance has been scant. In order to gain a better understanding of the potential utility of the antagonists in insect investigations, a conventional Hh antagonist, sonidegib, was used to evaluate the effects on the development of Drosophila larvae. The results showed that early instar larvae exposed to sonidegib exhibited new epidermal abnormalities and decreased motility after molting. Transcriptome analysis revealed that Sonidegib had a profound effect on chitin-based cuticle development throughout all stages of larvae. Physiological experiments revealed that sonidegib suppressed the epidermis formation and decreased the chitin content. The results of this study shed new light on the potential use of Hh antagonists in agricultural pest management.
Collapse
Affiliation(s)
- Zhiyan Jiang
- National Key Laboratory of Green Pesticide, Key Laboratory of Integrated Pest Management of Crop in South China, Ministry of Agriculture and Rural Affairs, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, PR China.
| | - Liwen Qu
- National Key Laboratory of Green Pesticide, Key Laboratory of Integrated Pest Management of Crop in South China, Ministry of Agriculture and Rural Affairs, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, PR China.
| | - Gaofeng Cui
- National Key Laboratory of Green Pesticide, Key Laboratory of Integrated Pest Management of Crop in South China, Ministry of Agriculture and Rural Affairs, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, PR China.
| | - Guohua Zhong
- National Key Laboratory of Green Pesticide, Key Laboratory of Integrated Pest Management of Crop in South China, Ministry of Agriculture and Rural Affairs, Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, PR China.
| |
Collapse
|
6
|
She R, Fair T, Schaefer NK, Saunders RA, Pavlovic BJ, Weissman JS, Pollen AA. Comparative landscape of genetic dependencies in human and chimpanzee stem cells. Cell 2023; 186:2977-2994.e23. [PMID: 37343560 PMCID: PMC10461406 DOI: 10.1016/j.cell.2023.05.043] [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: 08/31/2022] [Revised: 03/14/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023]
Abstract
Comparative studies of great apes provide a window into our evolutionary past, but the extent and identity of cellular differences that emerged during hominin evolution remain largely unexplored. We established a comparative loss-of-function approach to evaluate whether human cells exhibit distinct genetic dependencies. By performing genome-wide CRISPR interference screens in human and chimpanzee pluripotent stem cells, we identified 75 genes with species-specific effects on cellular proliferation. These genes comprised coherent processes, including cell-cycle progression and lysosomal signaling, which we determined to be human-derived by comparison with orangutan cells. Human-specific robustness to CDK2 and CCNE1 depletion persisted in neural progenitor cells and cerebral organoids, supporting the G1-phase length hypothesis as a potential evolutionary mechanism in human brain expansion. Our findings demonstrate that evolutionary changes in human cells reshaped the landscape of essential genes and establish a platform for systematically uncovering latent cellular and molecular differences between species.
Collapse
Affiliation(s)
- Richard She
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Tyler Fair
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Nathan K Schaefer
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Reuben A Saunders
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, CA, USA
| | - Bryan J Pavlovic
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan S Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute Technology, Cambridge, MA 02142, USA.
| | - Alex A Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
She R, Fair T, Schaefer NK, Saunders RA, Pavlovic BJ, Weissman JS, Pollen AA. Comparative landscape of genetic dependencies in human and chimpanzee stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.19.533346. [PMID: 36993685 PMCID: PMC10055274 DOI: 10.1101/2023.03.19.533346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Comparative studies of great apes provide a window into our evolutionary past, but the extent and identity of cellular differences that emerged during hominin evolution remain largely unexplored. We established a comparative loss-of-function approach to evaluate whether changes in human cells alter requirements for essential genes. By performing genome-wide CRISPR interference screens in human and chimpanzee pluripotent stem cells, we identified 75 genes with species-specific effects on cellular proliferation. These genes comprised coherent processes, including cell cycle progression and lysosomal signaling, which we determined to be human-derived by comparison with orangutan cells. Human-specific robustness to CDK2 and CCNE1 depletion persisted in neural progenitor cells, providing support for the G1-phase length hypothesis as a potential evolutionary mechanism in human brain expansion. Our findings demonstrate that evolutionary changes in human cells can reshape the landscape of essential genes and establish a platform for systematically uncovering latent cellular and molecular differences between species.
Collapse
Affiliation(s)
- Richard She
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- These authors contributed equally: Richard She, Tyler Fair
| | - Tyler Fair
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- These authors contributed equally: Richard She, Tyler Fair
| | - Nathan K. Schaefer
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Reuben A. Saunders
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, CA, USA
| | - Bryan J. Pavlovic
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan S. Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute Technology, Cambridge 02142, MA
| | - Alex A. Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Lead contact
| |
Collapse
|
8
|
Abstract
Hedgehog (Hh) proteins constitute one family of a small number of secreted signaling proteins that together regulate multiple aspects of animal development, tissue homeostasis and regeneration. Originally uncovered through genetic analyses in Drosophila, their subsequent discovery in vertebrates has provided a paradigm for the role of morphogens in positional specification. Most strikingly, the Sonic hedgehog protein was shown to mediate the activity of two classic embryonic organizing centers in vertebrates and subsequent studies have implicated it and its paralogs in a myriad of processes. Moreover, dysfunction of the signaling pathway has been shown to underlie numerous human congenital abnormalities and diseases, especially certain types of cancer. This review focusses on the genetic studies that uncovered the key components of the Hh signaling system and the subsequent, biochemical, cell and structural biology analyses of their functions. These studies have revealed several novel processes and principles, shedding new light on the cellular and molecular mechanisms underlying cell-cell communication. Notable amongst these are the involvement of cholesterol both in modifying the Hh proteins and in activating its transduction pathway, the role of cytonemes, filipodia-like extensions, in conveying Hh signals between cells; and the central importance of the Primary Cilium as a cellular compartment within which the components of the signaling pathway are sequestered and interact.
Collapse
Affiliation(s)
- Philip William Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
9
|
Lo M, Sharir A, Paul MD, Torosyan H, Agnew C, Li A, Neben C, Marangoni P, Xu L, Raleigh DR, Jura N, Klein OD. CNPY4 inhibits the Hedgehog pathway by modulating membrane sterol lipids. Nat Commun 2022; 13:2407. [PMID: 35504891 PMCID: PMC9065090 DOI: 10.1038/s41467-022-30186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/20/2022] [Indexed: 11/09/2022] Open
Abstract
The Hedgehog (HH) pathway is critical for development and adult tissue homeostasis. Aberrant HH signaling can lead to congenital malformations and diseases including cancer. Although cholesterol and several oxysterol lipids have been shown to play crucial roles in HH activation, the molecular mechanisms governing their regulation remain unresolved. Here, we identify Canopy4 (CNPY4), a Saposin-like protein, as a regulator of the HH pathway that modulates levels of membrane sterol lipids. Cnpy4-/- embryos exhibit multiple defects consistent with HH signaling perturbations, most notably changes in digit number. Knockdown of Cnpy4 hyperactivates the HH pathway in vitro and elevates membrane levels of accessible sterol lipids, such as cholesterol, an endogenous ligand involved in HH activation. Our data demonstrate that CNPY4 is a negative regulator that fine-tunes HH signal transduction, revealing a previously undescribed facet of HH pathway regulation that operates through control of membrane composition.
Collapse
Affiliation(s)
- Megan Lo
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Amnon Sharir
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Ein Kerem, Jerusalem, Israel
| | - Michael D Paul
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Hayarpi Torosyan
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Christopher Agnew
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Amy Li
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Cynthia Neben
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Pauline Marangoni
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94158, USA.
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA.
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA.
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA, USA.
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Makamte S, Thureau A, Jabrani A, Paquelin A, Plessis A, Sanial M, Rudenko O, Oteri F, Baaden M, Biou V. A large disordered region confers a wide spanning volume to vertebrate Suppressor of Fused as shown in a trans-species solution study. J Struct Biol 2022; 214:107853. [DOI: 10.1016/j.jsb.2022.107853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/10/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
11
|
Li W, Wang L, Wierbowski BM, Lu M, Dong F, Liu W, Li S, Wang P, Salic A, Gong X. Structural insights into proteolytic activation of the human Dispatched1 transporter for Hedgehog morphogen release. Nat Commun 2021; 12:6966. [PMID: 34845226 PMCID: PMC8630017 DOI: 10.1038/s41467-021-27257-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 11/08/2021] [Indexed: 12/26/2022] Open
Abstract
The membrane protein Dispatched (Disp), which belongs to the RND family of small molecule transporters, is essential for Hedgehog (Hh) signaling, by catalyzing the extracellular release of palmitate- and cholesterol-modified Hh ligands from producing cells. Disp function requires Furin-mediated proteolytic cleavage of its extracellular domain, but how this activates Disp remains obscure. Here, we employ cryo-electron microscopy to determine atomic structures of human Disp1 (hDisp1), before and after cleavage, and in complex with lipid-modified Sonic hedgehog (Shh) ligand. These structures, together with biochemical data, reveal that proteolytic cleavage opens the extracellular domain of hDisp1, removing steric hindrance to Shh binding. Structure-guided functional experiments demonstrate the role of hDisp1-Shh interactions in ligand release. Our results clarify the mechanisms of hDisp1 activation and Shh morphogen release, and highlight how a unique proteolytic cleavage event enabled acquisition of a protein substrate by a member of a family of small molecule transporters.
Collapse
Affiliation(s)
- Wanqiu Li
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China ,grid.263817.90000 0004 1773 1790Present Address: Department of Pharmacology, School of Medicine, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Linlin Wang
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Bradley M. Wierbowski
- grid.38142.3c000000041936754XDepartment of Cell Biology, Harvard Medical School, Boston, MA 02115 USA
| | - Mo Lu
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Feitong Dong
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Wenchen Liu
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Sisi Li
- grid.263817.90000 0004 1773 1790Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China ,grid.508211.f0000 0004 6004 3854Present Address: Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, 518060 Shenzhen, Guangdong China
| | - Peiyi Wang
- grid.263817.90000 0004 1773 1790SUSTech Cryo-EM Facility Center, Southern University of Science and Technology, 518055 Shenzhen, Guangdong China
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Xin Gong
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| |
Collapse
|
12
|
Kinnebrew M, Luchetti G, Sircar R, Frigui S, Viti LV, Naito T, Beckert F, Saheki Y, Siebold C, Radhakrishnan A, Rohatgi R. Patched 1 reduces the accessibility of cholesterol in the outer leaflet of membranes. eLife 2021; 10:e70504. [PMID: 34698632 PMCID: PMC8654371 DOI: 10.7554/elife.70504] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
A long-standing mystery in vertebrate Hedgehog signaling is how Patched 1 (PTCH1), the receptor for Hedgehog ligands, inhibits the activity of Smoothened, the protein that transmits the signal across the membrane. We previously proposed (Kinnebrew et al., 2019) that PTCH1 inhibits Smoothened by depleting accessible cholesterol from the ciliary membrane. Using a new imaging-based assay to directly measure the transport activity of PTCH1, we find that PTCH1 depletes accessible cholesterol from the outer leaflet of the plasma membrane. This transport activity is terminated by binding of Hedgehog ligands to PTCH1 or by dissipation of the transmembrane potassium gradient. These results point to the unexpected model that PTCH1 moves cholesterol from the outer to the inner leaflet of the membrane in exchange for potassium ion export in the opposite direction. Our study provides a plausible solution for how PTCH1 inhibits SMO by changing the organization of cholesterol in membranes and establishes a general framework for studying how proteins change cholesterol accessibility to regulate membrane-dependent processes in cells.
Collapse
Affiliation(s)
- Maia Kinnebrew
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| | - Giovanni Luchetti
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
- Department of Physiological Chemistry, GenentechSouth San FranciscoUnited States
| | - Ria Sircar
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| | - Sara Frigui
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| | - Lucrezia Vittoria Viti
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Tomoki Naito
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
| | - Francis Beckert
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| | - Yasunori Saheki
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Rajat Rohatgi
- Department of Biochemistry and Medicine, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
13
|
Xue Z, Wang W, Shen J, Zhang J, Zhang X, Liu X. A Patched-Like Protein PsPTL Is Not Essential for the Growth and Response to Various Stresses in Phytophthora sojae. Front Microbiol 2021; 12:673784. [PMID: 34690942 PMCID: PMC8530017 DOI: 10.3389/fmicb.2021.673784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 09/07/2021] [Indexed: 11/18/2022] Open
Abstract
Patched (Ptc) and Patched-related (Ptr) proteins containing sterol-sensing domains (SSD) and Patched domains are highly conserved in eukaryotes for lipid transport and metabolism. Four proteins containing predicted SSD and Patched domains were simultaneously found by searching the Phytophthora sojae genome database, and one of them was identified as a Patched-like (PTL) protein. Here, we investigated the biological function of PsPTL. The expression level of PsPTL was higher during mycelial and sporulation stages, compared to zoospore (ZO), cyst, and germinated-cyst stages, without significant change during infection. However, deletion of PsPTL using CRISPR/Cas9 had no significant effect on the growth, development, or virulence of P. sojae. Further investigations showed that PsPTL is not essential for P. sojae to cope with external stresses such as temperature, pH, oxidative and osmotic pressure. In addition, this gene did not appear to play an essential role in P. sojae’s response to exogenous sterols. The transcript levels of the other three proteins containing predicted SSD and Patched domains were also not significantly upregulated in PsPTL deletion transformants. Our studies demonstrated that PsPTL is not an essential protein for P. sojae under the tested conditions, and more in-depth research is required for revealing the potential functions of PsPTL under special conditions or in other signaling pathways.
Collapse
Affiliation(s)
- Zhaolin Xue
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing, China
| | - Weizhen Wang
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jinghuan Shen
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jinhui Zhang
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Xitao Zhang
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, China
| | - Xili Liu
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing, China.,State Key Laboratory of Crop Stress Biology for Arid Areas, College of Plant Protection, Northwest A&F University, Yangling, China
| |
Collapse
|
14
|
Astrocyte-Derived Extracellular Vesicle-Mediated Activation of Primary Ciliary Signaling Contributes to the Development of Morphine Tolerance. Biol Psychiatry 2021; 90:575-585. [PMID: 34417054 DOI: 10.1016/j.biopsych.2021.06.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/17/2021] [Accepted: 06/07/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Morphine is used extensively in the clinical setting owing to its beneficial effects, such as pain relief; its therapeutic utility is limited because the prolonged use of morphine often results in tolerance and addiction. Astrocytes in the brain are a direct target of morphine action and play an essential role in the development of morphine tolerance. Primary cilia and the cilia-mediated sonic hedgehog (SHH) signaling pathways have been shown to play a role in drug resistance and morphine tolerance, respectively. Extracellular vesicles (EVs) play important roles as cargo-carrying vesicles mediating communication among cells and tissues. METHODS C57BL/6N mice were administered morphine for 8 days to develop tolerance, which was determined using the tail-flick and hot plate assays. EVs were separated from astrocyte-conditioned media using either size exclusion chromatography or ultracentrifugation approaches, followed by characterization of EVs using nanoparticle tracking analysis for EV size distribution and number, Western blotting for EV markers, and electron microscopy for EV morphology. Astrocytes were treated with EVs for 24 hours, followed by assessing primary cilia by fluorescent immunostaining for primary cilia markers (ARL13B and acetylated tubulin). RESULTS Morphine-tolerant mice exhibited an increase in primary cilia length and percentage of ciliated astrocytes. The levels of SHH protein were upregulated in morphine-stimulated astrocyte-derived EVs. SHH on morphine-stimulated astrocyte-derived EVs activated SHH signaling in astrocytes through primary cilia. Our in vivo study demonstrated that inhibition of either EV release or primary cilia prevents morphine tolerance in mice. CONCLUSIONS EV-mediated primary ciliogenesis contributes to the development of morphine tolerance.
Collapse
|
15
|
Yang S, Zhang Y, Yang C, Wu X, El Oud SM, Chen R, Cai X, Wu XS, Lan G, Zheng X. Competitive coordination of the dual roles of the Hedgehog co-receptor in homophilic adhesion and signal reception. eLife 2021; 10:65770. [PMID: 34003115 PMCID: PMC8131103 DOI: 10.7554/elife.65770] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
Hedgehog (Hh) signaling patterns embryonic tissues and contributes to homeostasis in adults. In Drosophila, Hh transport and signaling are thought to occur along a specialized class of actin-rich filopodia, termed cytonemes. Here, we report that Interference hedgehog (Ihog) not only forms a Hh receptor complex with Patched to mediate intracellular signaling, but Ihog also engages in trans-homophilic binding leading to cytoneme stabilization in a manner independent of its role as the Hh receptor. Both functions of Ihog (trans-homophilic binding for cytoneme stabilization and Hh binding for ligand sensing) involve a heparin-binding site on the first fibronectin repeat of the extracellular domain. Thus, the Ihog-Ihog interaction and the Hh-Ihog interaction cannot occur simultaneously for a single Ihog molecule. By combining experimental data and mathematical modeling, we determined that Hh-Ihog heterophilic interaction dominates and Hh can disrupt and displace Ihog molecules involved in trans-homophilic binding. Consequently, we proposed that the weaker Ihog-Ihog trans interaction promotes and stabilizes direct membrane contacts along cytonemes and that, as the cytoneme encounters secreted Hh ligands, the ligands trigger release of Ihog from trans Ihog-Ihog complex enabling transport or internalization of the Hh ligand-Ihog-Patched -receptor complex. Thus, the seemingly incompatible functions of Ihog in homophilic adhesion and ligand binding cooperate to assist Hh transport and reception along the cytonemes.
Collapse
Affiliation(s)
- Shu Yang
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Ya Zhang
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Chuxuan Yang
- Department of Physics, George Washington University, Washington, United States
| | - Xuefeng Wu
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Sarah Maria El Oud
- Department of Physics, George Washington University, Washington, United States
| | - Rongfang Chen
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Xudong Cai
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Xufeng S Wu
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Ganhui Lan
- Department of Physics, George Washington University, Washington, United States
| | - Xiaoyan Zheng
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| |
Collapse
|
16
|
Emmons-Bell M, Hariharan IK. Membrane potential regulates Hedgehog signalling in the Drosophila wing imaginal disc. EMBO Rep 2021; 22:e51861. [PMID: 33629503 PMCID: PMC8024891 DOI: 10.15252/embr.202051861] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/29/2020] [Accepted: 01/15/2021] [Indexed: 01/02/2023] Open
Abstract
While the membrane potential of cells has been shown to be patterned in some tissues, specific roles for membrane potential in regulating signalling pathways that function during development are still being established. In the Drosophila wing imaginal disc, Hedgehog (Hh) from posterior cells activates a signalling pathway in anterior cells near the boundary which is necessary for boundary maintenance. Here, we show that membrane potential is patterned in the wing disc. Anterior cells near the boundary, where Hh signalling is most active, are more depolarized than posterior cells across the boundary. Elevated expression of the ENaC channel Ripped Pocket (Rpk), observed in these anterior cells, requires Hh. Antagonizing Rpk reduces depolarization and Hh signal transduction. Using genetic and optogenetic manipulations, in both the wing disc and the salivary gland, we show that membrane depolarization promotes membrane localization of Smoothened and augments Hh signalling, independently of Patched. Thus, membrane depolarization and Hh‐dependent signalling mutually reinforce each other in cells immediately anterior to the compartment boundary.
Collapse
Affiliation(s)
- Maya Emmons-Bell
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
17
|
Reconstitution of Morphogen Signaling Gradients in Cultured Cells. Methods Mol Biol 2020. [PMID: 33340353 DOI: 10.1007/978-1-0716-1174-6_4] [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
Development of multicellular organisms depends on the proper establishment of signaling information in space and time. Secreted molecules called morphogens form concentration gradients in space and provide positional information to differentiating cells within the organism. Although the key molecular components of morphogen pathways have been identified, how the architectures and key parameters of morphogen pathways control the properties of signaling gradients, such as their size, speed, and robustness to perturbations, remains challenging to study in developing embryos. Reconstituting morphogen gradients in cell culture provides an alternative approach to address this question. Here we describe the methodology for reconstituting Sonic Hedgehog (SHH) signaling gradients in mouse fibroblast cells. The protocol includes the design of morphogen sending and receiving cell lines, the setup of radial and linear gradients, the quantitative time-lapse imaging, and the data analysis. Similar approaches could potentially be applied to other cell-cell communication pathways.
Collapse
|
18
|
Wang J, Dahmann C. Establishing compartment boundaries in Drosophila wing imaginal discs: An interplay between selector genes, signaling pathways and cell mechanics. Semin Cell Dev Biol 2020; 107:161-169. [PMID: 32732129 DOI: 10.1016/j.semcdb.2020.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 01/02/2023]
Abstract
The partitioning of cells into groups or 'compartments' separated by straight and sharp boundaries is important for tissue formation in animal development. Cells from neighboring compartments are characterized by distinct fates and functions and their continuous separation at compartment boundaries maintains proper tissue organization. Signaling across compartment boundaries can induce the local expression of morphogens that in turn direct growth and patterning of the surrounding cells. Compartment boundaries play therefore an important role in tissue development. Compartment boundaries were first identified in the early 1970s in the Drosophila wing. Here, we review the role of compartment boundaries in growth and patterning of the developing wing and then discuss the genetic and physical mechanisms underlying cell separation at compartment boundaries in this tissue.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
| | - Christian Dahmann
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
19
|
Jiang W, Yao X, Shan Z, Li W, Gao Y, Zhang Q. E3 ligase Herc4 regulates Hedgehog signalling through promoting Smoothened degradation. J Mol Cell Biol 2020; 11:791-803. [PMID: 30925584 PMCID: PMC7261483 DOI: 10.1093/jmcb/mjz024] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/24/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022] Open
Abstract
Hedgehog (Hh) signalling plays conserved roles in controlling embryonic development; its dysregulation causes many diseases including cancers. The G protein-coupled receptor Smoothened (Smo) is the key signal transducer of the Hh pathway, whose posttranslational regulation has been shown to be critical for its accumulation and activation. Ubiquitination has been reported an essential posttranslational regulation of Smo. Here, we identify a novel E3 ligase of Smo, Herc4, which binds to Smo, and regulates Hh signalling by controlling Smo ubiquitination and degradation. Interestingly, our data suggest that Herc4-mediated Smo degradation is regulated by Hh in PKA-primed phosphorylation-dependent and independent manners.
Collapse
Affiliation(s)
- Weirong Jiang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xia Yao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Zhaoliang Shan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Wenting Li
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Yuxue Gao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Qing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, China
| |
Collapse
|
20
|
Chen Y, Yu H, Pask AJ, Fujiyama A, Suzuki Y, Sugano S, Shaw G, Renfree MB. Hormone-responsive genes in the SHH and WNT/β-catenin signaling pathways influence urethral closure and phallus growth. Biol Reprod 2019; 99:806-816. [PMID: 29767687 DOI: 10.1093/biolre/ioy117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 05/13/2018] [Indexed: 11/14/2022] Open
Abstract
Environmental endocrine disruptors (EEDs) that affect androgen or estrogen activity may disrupt gene regulation during phallus development to cause hypospadias or a masculinized clitoris. We treated developing male tammar wallabies with estrogen and females with androgen from day 20-40 postpartum (pp) during the androgen imprinting window of sensitivity. Estrogen inhibited phallus elongation but had no effect on urethral closure and did not significantly depress testicular androgen synthesis. Androgen treatment in females did not promote phallus elongation but initiated urethral closure. Phalluses were collected for transcriptome sequencing at day 50 pp when they first become sexually dimorphic to examine changes in two signaling pathways, sonic hedgehog (SHH) and wingless-type MMTV integration site family (WNT)/β-catenin. SHH mRNA and β-catenin were predominantly expressed in the urethral epithelium in the tammar phallus, as in eutherian mammals. Estrogen treatment and castration of males induced an upregulation of SHH, while androgen treatment downregulated SHH. These effects appear to be direct since we detected putative estrogen receptor α (ERα) and androgen receptor (AR) binding sites near SHH. WNT5A, like SHH, was downregulated by androgen, while WNT4 was upregulated in female phalluses after androgen treatment. After estrogen treatment, WIF1 and WNT7A were both downregulated in male phalluses. After castration, WNT9A was upregulated. These results suggest that SHH and WNT pathways are regulated by both estrogen and androgen to direct the proliferation and elongation of the phallus during differentiation. Their response to exogenous hormones makes these genes potential targets of EEDs in the etiology of abnormal phallus development including hypospadias.
Collapse
Affiliation(s)
- Yu Chen
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Hongshi Yu
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Asao Fujiyama
- Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Sumio Sugano
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Geoff Shaw
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Zhang X, Feng L, Qiao N, Liu Y, Zhang DC, Yin H. Cloning, expression pattern and functional characterization of fused, an important kinase of the Hedgehog signalling pathway from Locusta migratoria(Orthoptera: Acridoidea). BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1637781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Xiaohong Zhang
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Li Feng
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Ning Qiao
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Yachao Liu
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Dao Chuan Zhang
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Hong Yin
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| |
Collapse
|
22
|
Giordano C, Ruel L, Poux C, Therond P. Protein association changes in the Hedgehog signaling complex mediate differential signaling strength. Development 2018; 145:145/24/dev166850. [PMID: 30541874 DOI: 10.1242/dev.166850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 11/07/2018] [Indexed: 01/20/2023]
Abstract
Hedgehog (Hh) is a conserved morphogen that controls cell differentiation and tissue patterning in metazoans. In Drosophila, the Hh signal is transduced from the G protein-coupled receptor Smoothened (Smo) to the cytoplasmic Hh signaling complex (HSC). How activated Smo is translated into a graded activation of the downstream pathway is still not well understood. In this study, we show that the last amino acids of the cytoplasmic tail of Smo, in combination with G protein-coupled receptor kinase 2 (Gprk2), bind to the regulatory domain of Fused (Fu) and highly activate its kinase activity. We further show that this binding induces changes in the association of Fu protein with the HSC and increases the proximity of the Fu catalytic domain to its substrate, the Costal2 kinesin. We propose a new model in which, depending on the magnitude of Hh signaling, Smo and Gprk2 modulate protein association and conformational changes in the HSC, which are responsible for the differential activation of the pathway.
Collapse
Affiliation(s)
- Cecile Giordano
- Université Côte d'Azur, CNRS, Inserm, iBV, 06108 Nice, France
| | - Laurent Ruel
- Université Côte d'Azur, CNRS, Inserm, iBV, 06108 Nice, France
| | - Candice Poux
- Stockholms Universitet, Wenner-Grens Institut, SE-106 91 Stockholm, Sweden
| | - Pascal Therond
- Université Côte d'Azur, CNRS, Inserm, iBV, 06108 Nice, France
| |
Collapse
|
23
|
Liu A. Proteostasis in the Hedgehog signaling pathway. Semin Cell Dev Biol 2018; 93:153-163. [PMID: 31429406 DOI: 10.1016/j.semcdb.2018.10.009] [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: 07/13/2018] [Revised: 10/11/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022]
Abstract
The Hedgehog (Hh) signaling pathway is crucial for the development of vertebrate and invertebrate animals alike. Hh ligand binds its receptor Patched (Ptc), allowing the activation of the obligate signal transducer Smoothened (Smo). The levels and localizations of both Ptc and Smo are regulated by ubiquitination, and Smo is under additional regulation by phosphorylation and SUMOylation. Downstream of Smo, the Ci/Gli family of transcription factors regulates the transcriptional responses to Hh. Phosphorylation, ubiquitination and SUMOylation are important for the stability and localization of Ci/Gli proteins and Hh signaling output. Finally, Suppressor of Fused directly regulates Ci/Gli proteins and itself is under proteolytic regulation that is critical for normal Hh signaling.
Collapse
Affiliation(s)
- Aimin Liu
- Department of Biology, Eberly College of Science, Huck Institute of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, United States.
| |
Collapse
|
24
|
Abstract
First described in Drosophila, Hedgehog signalling is a key regulator of embryonic development and tissue homeostasis and its dysfunction underlies a variety of human congenital anomalies and diseases. Although now recognised as a major target for cancer therapy as well as a mediator of directed stem cell differentiation, the unveiling of the function and mechanisms of Hedgehog signalling was driven largely by an interest in basic developmental biology rather than clinical need. Here, I describe how curiosity about embryonic patterning led to the identification of the family of Hedgehog signalling proteins and the pathway that transduces their activity, and ultimately to the development of drugs that block this pathway.
Collapse
Affiliation(s)
- Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| |
Collapse
|
25
|
Gong X, Qian H, Cao P, Zhao X, Zhou Q, Lei J, Yan N. Structural basis for the recognition of Sonic Hedgehog by human Patched1. Science 2018; 361:science.aas8935. [DOI: 10.1126/science.aas8935] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/19/2018] [Indexed: 12/21/2022]
|
26
|
RND transporters in the living world. Res Microbiol 2018; 169:363-371. [PMID: 29577985 DOI: 10.1016/j.resmic.2018.03.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 02/27/2018] [Accepted: 03/01/2018] [Indexed: 12/22/2022]
Abstract
Transporters of the RND superfamily are well-known as the major drug efflux pumps of Gram-negative bacteria. However, they are widespread in organisms ranging from Archaea to Eukaryotes, and perform diverse functions. This review gives a brief overview of these diverse members of the superfamily with emphasis on their structure and functions.
Collapse
|
27
|
Alonso N, Cañueto J, Ciria S, Bueno E, Palacios-Alvarez I, Alegre M, Badenas C, Barreiro A, Pena L, Maldonado C, Nespeira-Jato MV, Peña-Penabad C, Azon A, Gavrilova M, Ferrer I, Sanmartin O, Robles L, Hernandez-Martin A, Urioste M, Puig S, Puig L, Gonzalez-Sarmiento R. Novel clinical and molecular findings in Spanish patients with naevoid basal cell carcinoma syndrome. Br J Dermatol 2017; 178:198-206. [PMID: 28733979 DOI: 10.1111/bjd.15835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Naevoid basal cell carcinoma syndrome (NBCCS) is an autosomal dominant disorder characterized by developmental alterations and multiple basal cell carcinomas. Mutations in PTCH1, which encodes a membrane receptor for Sonic Hedgehog, are associated with the development of the disease. Most of them produce a truncated protein, which is unable to suppress Smoothened protein and continuously activates the downstream pathway. OBJECTIVES We aimed to characterize 22 unrelated Spanish patients with NBCCS, the largest cohort with Gorlin syndrome reported to date in Spain. METHODS Genomic analysis of PTCH1 was performed in patients with NBCCS and controls, and mutations were analysed using bioinformatics tools. RESULTS We report for the first time two young patients, one each with uterus didelphys and ganglioneuroma, within the context of NBCCS. One patient showing a severe phenotype of the disease had developed basal cell carcinomas since childhood. Sanger sequencing of PTCH1 in this cohort identified 17 novel truncating mutations (11 frameshift, five nonsense and one mutation affecting an exon-intron splice site) and two novel missense mutations that were predicted to be pathogenic. The patients showed great clinical variability and inconsistent genotype-phenotype correlation, as seen in relatives carrying similar mutations. CONCLUSIONS This study contributes to increase the pool of clinical manifestations of NBCCS, as well as increasing the number of pathogenic mutations identified in PTCH1 predisposing to the condition. The inconsistencies found between phenotype and genotype suggest the involvement of other modifying factors, genetic, epigenetic or environmental.
Collapse
Affiliation(s)
- N Alonso
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain.,Rheumatology and Bone Disease Unit, Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, U.K
| | - J Cañueto
- Department of Dermatology, Salamanca University Hospital, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Institute of Molecular and Cellular Biology of Cancer (IBMCC), University Hospital of Salamanca, University of Salamanca-CSIC, Salamanca, Spain
| | - S Ciria
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - E Bueno
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Institute of Molecular and Cellular Biology of Cancer (IBMCC), University Hospital of Salamanca, University of Salamanca-CSIC, Salamanca, Spain
| | | | - M Alegre
- Department of Dermatology, Hospital Santa Creu i San Pau, Barcelona, Spain
| | - C Badenas
- Biochemistry and Molecular Genetics, Melanoma Unit, Hospital Clinic i Provincial, Barcelona, Spain.,Institut de Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Barcelona, Spain
| | - A Barreiro
- Department of Dermatology, Melanoma Unit, Hospital Clinic i Provincial, Barcelona, Spain
| | - L Pena
- Familial Cancer Clinical Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - C Maldonado
- Department of Dermatology, Hospital Central de Asturias, Oviedo, Spain
| | - M V Nespeira-Jato
- Department of Dermatology, Hospital Universitario de La Coruña, La Coruña, Spain
| | - C Peña-Penabad
- Department of Dermatology, Hospital Universitario de La Coruña, La Coruña, Spain
| | - A Azon
- Department of Dermatology, Hospital San Joan de Reus, Reus, Spain
| | - M Gavrilova
- Department of Dermatology, Hospital Clínico de Valencia, Valencia, Spain
| | - I Ferrer
- Department of Dermatology, Hospital General Universitario de Valencia, Valencia, Spain
| | - O Sanmartin
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain
| | - L Robles
- Hereditary Cancer Unit, Hospital 12 de Octubre, Madrid, Spain
| | | | - M Urioste
- Familial Cancer Clinical Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - S Puig
- Institut de Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Barcelona, Spain.,Department of Dermatology, Melanoma Unit, Hospital Clinic i Provincial, Barcelona, Spain
| | - L Puig
- Department of Dermatology, Hospital Santa Creu i San Pau, Barcelona, Spain
| | - R Gonzalez-Sarmiento
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Institute of Molecular and Cellular Biology of Cancer (IBMCC), University Hospital of Salamanca, University of Salamanca-CSIC, Salamanca, Spain
| |
Collapse
|
28
|
Singh R, Lauth M. Emerging Roles of DYRK Kinases in Embryogenesis and Hedgehog Pathway Control. J Dev Biol 2017; 5:E13. [PMID: 29615569 PMCID: PMC5831797 DOI: 10.3390/jdb5040013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 11/17/2017] [Accepted: 11/18/2017] [Indexed: 12/19/2022] Open
Abstract
Hedgehog (Hh)/GLI signaling is an important instructive cue in various processes during embryonic development, such as tissue patterning, stem cell maintenance, and cell differentiation. It also plays crucial roles in the development of many pediatric and adult malignancies. Understanding the molecular mechanisms of pathway regulation is therefore of high interest. Dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) comprise a group of protein kinases which are emerging modulators of signal transduction, cell proliferation, survival, and cell differentiation. Work from the last years has identified a close regulatory connection between DYRKs and the Hh signaling system. In this manuscript, we outline the mechanistic influence of DYRK kinases on Hh signaling with a focus on the mammalian situation. We furthermore aim to bring together what is known about the functional consequences of a DYRK-Hh cross-talk and how this might affect cellular processes in development, physiology, and pathology.
Collapse
Affiliation(s)
- Rajeev Singh
- Philipps University Marburg, Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor and Immune Biology (ZTI), Hans-Meerwein-Str. 3, 35043 Marburg, Germany.
| | - Matthias Lauth
- Philipps University Marburg, Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor and Immune Biology (ZTI), Hans-Meerwein-Str. 3, 35043 Marburg, Germany.
| |
Collapse
|
29
|
Sheybani-Deloui S, Chi L, Staite MV, Cain JE, Nieman BJ, Henkelman RM, Wainwright BJ, Potter SS, Bagli DJ, Lorenzo AJ, Rosenblum ND. Activated Hedgehog-GLI Signaling Causes Congenital Ureteropelvic Junction Obstruction. J Am Soc Nephrol 2017; 29:532-544. [PMID: 29109083 DOI: 10.1681/asn.2017050482] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/05/2017] [Indexed: 12/19/2022] Open
Abstract
Intrinsic ureteropelvic junction obstruction is the most common cause of congenital hydronephrosis, yet the underlying pathogenesis is undefined. Hedgehog proteins control morphogenesis by promoting GLI-dependent transcriptional activation and inhibiting the formation of the GLI3 transcriptional repressor. Hedgehog regulates differentiation and proliferation of ureteric smooth muscle progenitor cells during murine kidney-ureter development. Histopathologic findings of smooth muscle cell hypertrophy and stroma-like cells, consistently observed in obstructing tissue at the time of surgical correction, suggest that Hedgehog signaling is abnormally regulated during the genesis of congenital intrinsic ureteropelvic junction obstruction. Here, we demonstrate that constitutively active Hedgehog signaling in murine intermediate mesoderm-derived renal progenitors results in hydronephrosis and failure to develop a patent pelvic-ureteric junction. Tissue obstructing the ureteropelvic junction was marked as early as E13.5 by an ectopic population of cells expressing Ptch2, a Hedgehog signaling target. Constitutive expression of GLI3 repressor in Ptch1-deficient mice rescued ectopic Ptch2 expression and obstructive hydronephrosis. Whole transcriptome analysis of isolated Ptch2+ cells revealed coexpression of genes characteristic of stromal progenitor cells. Genetic lineage tracing indicated that stromal cells blocking the ureteropelvic junction were derived from intermediate mesoderm-derived renal progenitors and were distinct from the smooth muscle or epithelial lineages. Analysis of obstructive ureteric tissue resected from children with congenital intrinsic ureteropelvic junction obstruction revealed a molecular signature similar to that observed in Ptch1-deficient mice. Together, these results demonstrate a Hedgehog-dependent mechanism underlying mammalian intrinsic ureteropelvic junction obstruction.
Collapse
Affiliation(s)
| | - Lijun Chi
- Program in Developmental and Stem Cell Biology
| | - Marian V Staite
- Program in Developmental and Stem Cell Biology.,Departments of Physiology
| | | | - Brian J Nieman
- Program in Physiology and Experimental Medicine, and.,Medical Biophysics and Medical Imaging, and.,Mouse Imaging Centre, Toronto Centre for Phenogenomics Toronto, Ontario, Canada.,Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - R Mark Henkelman
- Medical Biophysics and Medical Imaging, and.,Mouse Imaging Centre, Toronto Centre for Phenogenomics Toronto, Ontario, Canada
| | - Brandon J Wainwright
- Genomics of Development and Disease Division, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and
| | - S Steven Potter
- Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Darius J Bagli
- Program in Developmental and Stem Cell Biology.,Departments of Physiology.,Division of Urology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Armando J Lorenzo
- Division of Urology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Norman D Rosenblum
- Program in Developmental and Stem Cell Biology, .,Departments of Physiology.,Division of Nephrology.,Paediatrics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Hedgehog mediated degradation of Ihog adhesion proteins modulates cell segregation in Drosophila wing imaginal discs. Nat Commun 2017; 8:1275. [PMID: 29097673 PMCID: PMC5668237 DOI: 10.1038/s41467-017-01364-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/12/2017] [Indexed: 11/08/2022] Open
Abstract
The Drosophila Hedgehog receptor functions to regulate the essential downstream pathway component, Smoothened, and to limit the range of signaling by sequestering Hedgehog protein signal within imaginal disc epithelium. Hedgehog receptor function requires both Patched and Ihog activity, the latter interchangeably encoded by interference hedgehog (ihog) or brother of ihog (boi). Here we show that Patched and Ihog activity are mutually required for receptor endocytosis and degradation, triggered by Hedgehog protein binding, and causing reduced levels of Ihog/Boi proteins in a stripe of cells at the anterior/posterior compartment boundary of the wing imaginal disc. This Ihog spatial discontinuity may contribute to classically defined cell segregation and lineage restriction at the anterior/posterior wing disc compartment boundary, as suggested by our observations that Ihog activity mediates aggregation of otherwise non-adherent cultured cells and that loss of Ihog activity disrupts wing disc cell segregation, even with downstream genetic rescue of Hedgehog signal response.
Collapse
|
31
|
Abstract
Communication between cells pervades the development and physiology of metazoans. In animals, this process is carried out by a relatively small number of signaling pathways, each consisting of a chain of biochemical events through which extracellular stimuli control the behavior of target cells. One such signaling system is the Hedgehog pathway, which is crucial in embryogenesis and is implicated in many birth defects and cancers. Although Hedgehog pathway components were identified by genetic analysis more than a decade ago, our understanding of the molecular mechanisms of signaling is far from complete. In this review, we focus on the biochemistry and cell biology of the Hedgehog pathway. We examine the unique biosynthesis of the Hedgehog ligand, its specialized release from cells into extracellular space, and the poorly understood mechanisms involved in ligand reception and pathway activation at the surface of target cells. We highlight several critical questions that remain open.
Collapse
Affiliation(s)
- Kostadin Petrov
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115; ,
| | - Bradley M Wierbowski
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115; ,
| | - Adrian Salic
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115; ,
| |
Collapse
|
32
|
Chen P, Zhou Z, Yao X, Pang S, Liu M, Jiang W, Jiang J, Zhang Q. Capping Enzyme mRNA-cap/RNGTT Regulates Hedgehog Pathway Activity by Antagonizing Protein Kinase A. Sci Rep 2017; 7:2891. [PMID: 28588207 PMCID: PMC5460166 DOI: 10.1038/s41598-017-03165-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 04/21/2017] [Indexed: 02/07/2023] Open
Abstract
Hedgehog (Hh) signaling plays a pivotal role in animal development and its deregulation in humans causes birth defects and several types of cancer. Protein Kinase A (PKA) modulates Hh signaling activity through phosphorylating the transcription factor Cubitus interruptus (Ci) and G protein coupled receptor (GPCR) family protein Smoothened (Smo) in Drosophila, but how PKA activity is regulated remains elusive. Here, we identify a novel regulator of the Hh pathway, the capping-enzyme mRNA-cap, which positively regulates Hh signaling activity through modulating PKA activity. We provide genetic and biochemical evidence that mRNA-cap inhibits PKA kinase activity to promote Hh signaling. Interestingly, regulation of Hh signaling by mRNA-cap depends on its cytoplasmic capping-enzyme activity. In addition, we show that the mammalian homolog of mRNA-cap, RNGTT, can replace mRNA-cap to play the same function in the Drosophila Hh pathway and that knockdown of Rngtt in cultured mammalian cells compromised Shh pathway activity, suggesting that RNGTT is functionally conserved. Our study makes an unexpected link between the mRNA capping machinery and the Hh signaling pathway, unveils a new facet of Hh signaling regulation, and reveals a potential drug target for modulating Hh signaling activity.
Collapse
Affiliation(s)
- Ping Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Zizhang Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Xia Yao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Shu Pang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Meijing Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Weirong Jiang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA. .,Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
| | - Qing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China.
| |
Collapse
|
33
|
Dickinson WJ, Yang Y, Schuske K, Akam M. CONSERVATION OF MOLECULAR PREPATTERNS DURING THE EVOLUTION OF CUTICLE MORPHOLOGY IN DROSOPHILA LARVAE. Evolution 2017; 47:1396-1406. [PMID: 28564885 DOI: 10.1111/j.1558-5646.1993.tb02162.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/1992] [Accepted: 12/29/1992] [Indexed: 11/29/2022]
Abstract
We are using patterns of cuticle specialization in Drosophila larvae as models to investigate the molecular, genetic, and developmental bases of morphological evolution. Members of the virilis species group differ markedly from one another in the distribution of hairs on the dorsal surface of first instar larvae. In particular, characteristic bands of hairs cover about 20% of each trunk segment in some species but about 70% in others. These major types do not correlate with recently proposed phylogenetic relationships, suggesting that similar phenotypes have arisen independently in different lineages. The patterns of expression of several genes that control or reflect intrasegmental patterning are indistinguishable in species with very different cuticle morphologies. We conclude that, in this case, morphology probably has evolved via altered response to a conserved molecular prepattern.
Collapse
Affiliation(s)
- W J Dickinson
- Department of Biology, University of Utah, Salt Lake City, Utah, 84112
| | | | - Kim Schuske
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, 94305-5020
| | - Michael Akam
- Wellcome/CRC Institute and Department of Genetics, University of Cambridge, Cambridge, CB2 1QR, UK
| |
Collapse
|
34
|
Moulton MJ, Letsou A. Modeling congenital disease and inborn errors of development in Drosophila melanogaster. Dis Model Mech 2016; 9:253-69. [PMID: 26935104 PMCID: PMC4826979 DOI: 10.1242/dmm.023564] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fly models that faithfully recapitulate various aspects of human disease and human health-related biology are being used for research into disease diagnosis and prevention. Established and new genetic strategies in Drosophila have yielded numerous substantial successes in modeling congenital disorders or inborn errors of human development, as well as neurodegenerative disease and cancer. Moreover, although our ability to generate sequence datasets continues to outpace our ability to analyze these datasets, the development of high-throughput analysis platforms in Drosophila has provided access through the bottleneck in the identification of disease gene candidates. In this Review, we describe both the traditional and newer methods that are facilitating the incorporation of Drosophila into the human disease discovery process, with a focus on the models that have enhanced our understanding of human developmental disorders and congenital disease. Enviable features of the Drosophila experimental system, which make it particularly useful in facilitating the much anticipated move from genotype to phenotype (understanding and predicting phenotypes directly from the primary DNA sequence), include its genetic tractability, the low cost for high-throughput discovery, and a genome and underlying biology that are highly evolutionarily conserved. In embracing the fly in the human disease-gene discovery process, we can expect to speed up and reduce the cost of this process, allowing experimental scales that are not feasible and/or would be too costly in higher eukaryotes.
Collapse
Affiliation(s)
- Matthew J Moulton
- Department of Human Genetics, University of Utah, 15 North 2030 East, Room 5100, Salt Lake City, UT 84112-5330, USA
| | - Anthea Letsou
- Department of Human Genetics, University of Utah, 15 North 2030 East, Room 5100, Salt Lake City, UT 84112-5330, USA
| |
Collapse
|
35
|
Simon E, Aguirre-Tamaral A, Aguilar G, Guerrero I. Perspectives on Intra- and Intercellular Trafficking of Hedgehog for Tissue Patterning. J Dev Biol 2016; 4:jdb4040034. [PMID: 29615597 PMCID: PMC5831803 DOI: 10.3390/jdb4040034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 12/14/2022] Open
Abstract
Intercellular communication is a fundamental process for correct tissue development. The mechanism of this process involves, among other things, the production and secretion of signaling molecules by specialized cell types and the capability of these signals to reach the target cells in order to trigger specific responses. Hedgehog (Hh) is one of the best-studied signaling pathways because of its importance during morphogenesis in many organisms. The Hh protein acts as a morphogen, activating its targets at a distance in a concentration-dependent manner. Post-translational modifications of Hh lead to a molecule covalently bond to two lipid moieties. These lipid modifications confer Hh high affinity to lipidic membranes, and intense studies have been carried out to explain its release into the extracellular matrix. This work reviews Hh molecule maturation, the intracellular recycling needed for its secretion and the proposed carriers to explain Hh transportation to the receiving cells. Special focus is placed on the role of specialized filopodia, also named cytonemes, in morphogen transport and gradient formation.
Collapse
Affiliation(s)
- Eléanor Simon
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Adrián Aguirre-Tamaral
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Gustavo Aguilar
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| | - Isabel Guerrero
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, CSIC-UAM, Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
36
|
Hyun J, Jung Y. MicroRNAs in liver fibrosis: Focusing on the interaction with hedgehog signaling. World J Gastroenterol 2016; 22:6652-6662. [PMID: 27547008 PMCID: PMC4970468 DOI: 10.3748/wjg.v22.i29.6652] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/08/2016] [Accepted: 06/29/2016] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is a repair process in response to damage in the liver; however, severe and chronic injury promotes the accumulation of fibrous matrix, destroying the normal functions and architecture of liver. Hepatic stellate cells (HSCs) are quiescent in normal livers, but in damaged livers, they transdifferentiate into myofibroblastic HSCs, which produce extracellular matrix proteins. Hedgehog (Hh) signaling orchestrates tissue reconstruction in damaged livers and contributes to liver fibrogenesis by regulating HSC activation. MicroRNAs (miRNAs), endogenous small non-coding RNAs interfering with RNA post-transcriptionally, regulate various cellular processes in healthy organisms. The dysregulation of miRNAs is closely associated with diseases, including liver diseases. Thus, miRNAs are good targets in the diagnosis and treatment of various diseases, including liver fibrosis; however, the regulatory mechanisms of miRNAs that interact with Hh signaling in liver fibrosis remain unclear. We review growing evidence showing the association of miRNAs with Hh signaling. Recent studies suggest that Hh-regulating miRNAs induce inactivation of HSCs, leading to decreased hepatic fibrosis. Although miRNA-delivery systems and further knowledge of interacting miRNAs with Hh signaling need to be improved for the clinical usage of miRNAs, recent findings indicate that the miRNAs regulating Hh signaling are promising therapeutic agents for treating liver fibrosis.
Collapse
|
37
|
Wieschaus E, Nüsslein-Volhard C. The Heidelberg Screen for Pattern Mutants of Drosophila: A Personal Account. Annu Rev Cell Dev Biol 2016; 32:1-46. [PMID: 27501451 DOI: 10.1146/annurev-cellbio-113015-023138] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In large-scale mutagenesis screens performed in 1979-1980 at the EMBL in Heidelberg, we isolated mutations affecting the pattern or structure of the larval cuticle in Drosophila. The 600 mutants we characterized could be assigned to 120 genes and represent the majority of such genes in the genome. These mutants subsequently provided a rich resource for understanding many fundamental developmental processes, such as the transcriptional hierarchies controlling segmentation, the establishment of cell states by signaling pathways, and the differentiation of epithelial cells. Most of the Heidelberg genes are now molecularly known, and many of them are conserved in other animals, including humans. Although the screens were initially driven entirely by curiosity, the mutants now serve as models for many human diseases. In this review, we describe the rationale of the screening procedures and provide a classification of the genes on the basis of their initial phenotypes and the subsequent molecular analyses.
Collapse
Affiliation(s)
- Eric Wieschaus
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544;
| | | |
Collapse
|
38
|
Structural Insights into the Niemann-Pick C1 (NPC1)-Mediated Cholesterol Transfer and Ebola Infection. Cell 2016; 165:1467-1478. [PMID: 27238017 PMCID: PMC7111323 DOI: 10.1016/j.cell.2016.05.022] [Citation(s) in RCA: 229] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/22/2016] [Accepted: 05/03/2016] [Indexed: 11/22/2022]
Abstract
Niemann-Pick disease type C (NPC) is associated with mutations in NPC1 and NPC2, whose gene products are key players in the endosomal/lysosomal egress of low-density lipoprotein-derived cholesterol. NPC1 is also the intracellular receptor for Ebola virus (EBOV). Here, we present a 4.4 Å structure of full-length human NPC1 and a low-resolution reconstruction of NPC1 in complex with the cleaved glycoprotein (GPcl) of EBOV, both determined by single-particle electron cryomicroscopy. NPC1 contains 13 transmembrane segments (TMs) and three distinct lumenal domains A (also designated NTD), C, and I. TMs 2–13 exhibit a typical resistance-nodulation-cell division fold, among which TMs 3–7 constitute the sterol-sensing domain conserved in several proteins involved in cholesterol metabolism and signaling. A trimeric EBOV-GPcl binds to one NPC1 monomer through the domain C. Our structural and biochemical characterizations provide an important framework for mechanistic understanding of NPC1-mediated intracellular cholesterol trafficking and Ebola virus infection. The cryo-EM structure of full-length human NPC1 was determined at 4.4 Å resolution Structure-guided biochemical analysis of cholesterol transfer from NPC2 to NPC1 Low-resolution cryo-EM structure of NPC1 bound to GPcl of Ebola virus was obtained A trimeric GPcl binds to one NPC1 through the crystal structure-revealed interface
Collapse
|
39
|
Christofi T, Apidianakis Y. Drosophila and the hallmarks of cancer. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2016; 135:79-110. [PMID: 23615878 DOI: 10.1007/10_2013_190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
: Cancer was the disease of the twentieth century. Today it is still a leading cause of death worldwide despite being intensively investigated. Abundant knowledge exists regarding the pathological and molecular mechanisms that drive healthy cells to become malignant and form metastatic tumors. The relation of oncogenes and tumor suppressors to the genetic trigger of carcinogenesis is unquestionable. However, the development of the disease requires many characteristics that due to their proven role in cancer are collectively described as the "hallmarks of cancer." We highlight here the historic discoveries made using the model organism Drosophila melanogaster and its contributions to biomedical and cancer research. Flies are utilized as a model organism for the investigation of each and every aspect of cancer hallmarks. Due to the significant conservation between flies and mammals at the signaling and tissue physiology level it is possible to explore the genes and mechanisms responsible for cancer pathogenesis in flies. Recent Drosophila studies suggest novel aspects of therapeutic intervention and are expected to guide cancer research in the twenty-first century.
Collapse
|
40
|
Ingham PW. Drosophila Segment Polarity Mutants and the Rediscovery of the Hedgehog Pathway Genes. Curr Top Dev Biol 2016; 116:477-88. [PMID: 26970635 DOI: 10.1016/bs.ctdb.2016.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The Nüsslein-Volhard and Wieschaus screen for mutations disrupting the segmentation of the Drosophila embryo revolutionized developmental genetics, leading the way to the identification of many of the transcription factors and signaling pathways that orchestrate development, not just in the fruit fly but across the animal kingdom. The Hedgehog signaling pathway is a case in point: yet remarkably, all but one of the genes encoding the Hedgehog pathway components-including Hedgehog itself-had previously been discovered, in some cases decades earlier. Here I review the original identification of these genes and consider why their significance remained obscure until the Nobel Prize winning screen.
Collapse
Affiliation(s)
- Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Department of Medicine, Imperial College, London, United Kingdom.
| |
Collapse
|
41
|
Srinath S, Iyengar AR, Mysorekar V. Sonic hedgehog in oral squamous cell carcinoma: An immunohistochemical study. J Oral Maxillofac Pathol 2016; 20:377-383. [PMID: 27721600 PMCID: PMC5051283 DOI: 10.4103/0973-029x.190906] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Recent studies have revealed the involvement of hedgehog (Hh) signaling component in proliferation and invasive behavior of many carcinomas. AIM This study aims to identify the expression of sonic Hh (SHH) protein of SHH pathway in oral epithelial dysplasia and oral squamous cell carcinoma (OSCC) using SHH (H-160) (Santa Cruz, sc-9042) which could have therapeutic implication in future. MATERIALS AND METHODS A total of 250 cases comprising 50 normal oral mucosa, 50 cases of oral epithelial dysplasia, 50 well, 50 moderate and 50 poorly differentiated OSCCs were included in the study. Immunohistochemical evaluation of SHH protein expression was conducted using monoclonal antibody. Interpretation of the expression was done by immunoreactive score of Remmele and Stegner (IRS) scoring method. STATISTICAL ANALYSIS Chi-Square test was used to analyze the results. RESULTS The study showed that SHH signaling molecules are highly expressed in OSCC, and their expression was mainly in the cytoplasm of epithelial cells. CONCLUSION The SHH signaling component is associated with the pathological parameter in OSCC and oral epithelial dysplasia.
Collapse
Affiliation(s)
- Sahana Srinath
- Department of Oral Pathology, GDCRI, Bengaluru, Karnataka, India
| | - Asha R Iyengar
- Department of Oral Medicine and Radiology, DAPMRV, Bengaluru, Karnataka, India
| | | |
Collapse
|
42
|
Bolatto C, Parada C, Revello F, Zuñiga A, Cabrera P, Cambiazo V. Spatial and temporal distribution of Patched-related protein in the Drosophila embryo. Gene Expr Patterns 2015; 19:120-8. [PMID: 26506022 DOI: 10.1016/j.gep.2015.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 10/08/2015] [Accepted: 10/21/2015] [Indexed: 12/20/2022]
Abstract
Patched-related (Ptr) encodes a protein with 12 potential transmembrane domains and a sterol-sensing domain that is closely related in predicted topology and domain organization to Patched, the canonical receptor of the Hedgehog pathway. Here we describe the production of an antibody specific for Drosophila Ptr and analyse its spatial and temporal distribution in the embryo. We find that at early developmental stages Ptr is predominantly localized at cell periphery but later on it becomes strongly and almost exclusively expressed in hemocytes. Interestingly Ptr null mutant embryos died without hatching. Our findings suggest that Ptr plays an essential function in Drosophila development, perhaps as a new receptor of embryonic hemocytes.
Collapse
Affiliation(s)
- Carmen Bolatto
- Laboratorio de Biología del Desarrollo, Departamento de Histología y Embriología, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay.
| | - Cristina Parada
- Laboratorio de Biología del Desarrollo, Departamento de Histología y Embriología, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay.
| | - Fiorella Revello
- Laboratorio de Biología del Desarrollo, Departamento de Histología y Embriología, Facultad de Medicina-Universidad de la República, Montevideo, Uruguay.
| | - Alejandro Zuñiga
- Laboratorio de Bioinformática y Expresión Génica, INTA-Universidad de Chile and Fondap Center for Genome Regulation (CGR), Santiago, Chile.
| | - Pablo Cabrera
- Laboratorio de Bioinformática y Expresión Génica, INTA-Universidad de Chile and Fondap Center for Genome Regulation (CGR), Santiago, Chile
| | - Verónica Cambiazo
- Laboratorio de Bioinformática y Expresión Génica, INTA-Universidad de Chile and Fondap Center for Genome Regulation (CGR), Santiago, Chile.
| |
Collapse
|
43
|
Lavore A, Pascual A, Salinas FM, Esponda-Behrens N, Martinez-Barnetche J, Rodriguez M, Rivera-Pomar R. Comparative analysis of zygotic developmental genes in Rhodnius prolixus genome shows conserved features on the tracheal developmental pathway. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 64:32-43. [PMID: 26187251 DOI: 10.1016/j.ibmb.2015.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 06/13/2015] [Accepted: 06/21/2015] [Indexed: 06/04/2023]
Abstract
Most of the in-depth studies on insect developmental genetic have been carried out in the fruit fly Drosophila melanogaster, an holometabolous insect, so much more still remains to be studied in hemimetabolous insects. Having Rhodnius prolixus sequenced genome available, we search for orthologue genes of zygotic signaling pathways, segmentation, and tracheogenesis in the R. prolixus genome and in three species of Triatoma genus transcriptomes, concluding that there is a high level of gene conservation. We also study the function of two genes required for tracheal system development in D. melanogaster - R. prolixus orthologues: trachealess (Rp-trh) and empty spiracles (Rp-ems). From that we see that Rp-trh is required for early tracheal development since Rp-trh RNAi shows that the primary tracheal branches fail to form. On the other hand, Rp-ems is implied in the proper formation of the posterior tracheal branches, in a similar way to D. melanogaster. These results represent the initial characterization of the genes involved in the tracheal development of an hemimetabolous insect building a bridge between the current genomic era and V. Wigglesworth's classical studies on insects' respiratory system physiology.
Collapse
Affiliation(s)
- A Lavore
- Centro de Bioinvestigaciones and Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Monteagudo 2772, 2700, Pergamino, Buenos Aires, Argentina.
| | - A Pascual
- Centro de Bioinvestigaciones and Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Monteagudo 2772, 2700, Pergamino, Buenos Aires, Argentina; Laboratorio de Genética y Genómica Funcional, Centro Regional de Estudios Genómicos, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bvd 120 y 62, La Plata, Buenos Aires, Argentina.
| | - F M Salinas
- Centro de Bioinvestigaciones and Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Monteagudo 2772, 2700, Pergamino, Buenos Aires, Argentina.
| | - N Esponda-Behrens
- Laboratorio de Genética y Genómica Funcional, Centro Regional de Estudios Genómicos, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bvd 120 y 62, La Plata, Buenos Aires, Argentina.
| | - J Martinez-Barnetche
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, Sta. María Ahuacatitlán, Cuernavaca, Mexico.
| | - M Rodriguez
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, Sta. María Ahuacatitlán, Cuernavaca, Mexico.
| | - R Rivera-Pomar
- Centro de Bioinvestigaciones and Departamento de Ciencias Básicas y Experimentales, Universidad Nacional del Noroeste de la Provincia de Buenos Aires, Monteagudo 2772, 2700, Pergamino, Buenos Aires, Argentina; Laboratorio de Genética y Genómica Funcional, Centro Regional de Estudios Genómicos, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Bvd 120 y 62, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
44
|
Ricolo D, Butí E, Araújo SJ. Drosophila melanogaster Hedgehog cooperates with Frazzled to guide axons through a non-canonical signalling pathway. Mech Dev 2015; 137:11-22. [DOI: 10.1016/j.mod.2015.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/13/2015] [Accepted: 04/27/2015] [Indexed: 12/16/2022]
|
45
|
Linder B, Weber S, Dittmann K, Adamski J, Hahn H, Uhmann A. A Functional and Putative Physiological Role of Calcitriol in Patched1/Smoothened Interaction. J Biol Chem 2015; 290:19614-28. [PMID: 26126827 DOI: 10.1074/jbc.m115.646141] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Indexed: 01/03/2023] Open
Abstract
The Patched1 (Ptch)-mediated inhibition of Smoothened (Smo) is still an open question. However, a direct Ptch/Smo interaction has been excluded, Smo modulators were identified, but the endogenous signal transmitting molecule remains undiscovered. Here, we demonstrate that calcitriol, the hormonally active form of vitamin D3, is an excellent candidate for transmission of Ptch/Smo interaction. Our study reveals that Ptch expression is sufficient to release calcitriol from the cell and that calcitriol inhibits Smo action and ciliary translocation by acting on a site distinct from the 7-transmembrane domain or the cysteine-rich domain. Moreover calcitriol strongly synergizes with itraconazole (ITZ) in Smo inhibition, which did not result from elevated calcitriol bioavailability due to ITZ-mediated 24-hydroxylase inhibition but rather from a direct interaction of the compounds at the level of Smo. Together, we suggest that calcitriol represents a possible endogenous transmitter of Ptch/Smo interaction. Moreover calcitriol or calcitriol derivatives combined with ITZ might be a treatment option of Hedgehog-associated cancers.
Collapse
Affiliation(s)
- Benedikt Linder
- From the Institute of Human Genetics, Tumor Genetics Group, and
| | - Susanne Weber
- the Department Genome Analysis Centre, Institute for Experimental Genetics, Helmholtz Zentrum Muenchen, National Research Center for Environment and Health, 85764 Neuherberg, Germany
| | - Kai Dittmann
- the Institute of Cellular and Molecular Immunology, University Medical Center, 37073 Goettingen and
| | - Jerzy Adamski
- the Department Genome Analysis Centre, Institute for Experimental Genetics, Helmholtz Zentrum Muenchen, National Research Center for Environment and Health, 85764 Neuherberg, Germany
| | - Heidi Hahn
- From the Institute of Human Genetics, Tumor Genetics Group, and
| | - Anja Uhmann
- From the Institute of Human Genetics, Tumor Genetics Group, and
| |
Collapse
|
46
|
Abstract
The Hedgehog (Hh) signalling pathway plays many important roles in development, homeostasis and tumorigenesis. The critical function of Hh signalling in bone formation has been identified in the past two decades. Here, we review the evolutionarily conserved Hh signalling mechanisms with an emphasis on the functions of the Hh signalling pathway in bone development, homeostasis and diseases. In the early stages of embryonic limb development, Sonic Hedgehog (Shh) acts as a major morphogen in patterning the limb buds. Indian Hedgehog (Ihh) has an essential function in endochondral ossification and induces osteoblast differentiation in the perichondrium. Hh signalling is also involved intramembrane ossification. Interactions between Hh and Wnt signalling regulate cartilage development, endochondral bone formation and synovial joint formation. Hh also plays an important role in bone homeostasis, and reducing Hh signalling protects against age-related bone loss. Disruption of Hh signalling regulation leads to multiple bone diseases, such as progressive osseous heteroplasia. Therefore, understanding the signalling mechanisms and functions of Hh signalling in bone development, homeostasis and diseases will provide important insights into bone disease prevention, diagnoses and therapeutics.
Collapse
|
47
|
Zhulyn O, Nieuwenhuis E, Liu YC, Angers S, Hui CC. Ptch2 shares overlapping functions with Ptch1 in Smo regulation and limb development. Dev Biol 2015; 397:191-202. [DOI: 10.1016/j.ydbio.2014.10.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 10/04/2014] [Accepted: 10/27/2014] [Indexed: 10/24/2022]
|
48
|
CHEN JIASHOU, LIU GUOZHONG, WENG XIAPING, LIU FAYUAN, LIN PINGDONG, LI HUITING, CHEN WENLIE, HUANG YUNMEI, LIU XIANXIANG, YE HONGZHI, LI XIHAI. Tougu Xiaotong formula induces chondrogenic differentiation in association with transforming growth factor-β1 and promotes proliferation in bone marrow stromal cells. Int J Mol Med 2014; 35:747-54. [DOI: 10.3892/ijmm.2014.2049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 12/08/2014] [Indexed: 11/06/2022] Open
|
49
|
Ranieri N, Thérond PP, Ruel L. Switch of PKA substrates from Cubitus interruptus to Smoothened in the Hedgehog signalosome complex. Nat Commun 2014; 5:5034. [PMID: 25289679 DOI: 10.1038/ncomms6034] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 08/20/2014] [Indexed: 12/20/2022] Open
Abstract
Hedgehog (Hh) signalling is crucial for developmental patterning and tissue homeostasis. In Drosophila, Hh signalling is mediated by a bifunctional transcriptional mediator, called Cubitus interruptus (Ci). Protein Kinase A (PKA)-dependent phosphorylation of the serpentine protein Smoothened (Smo) leads to Ci activation, whereas PKA-dependent phosphorylation of Ci leads to the formation of Ci repressor form. The mechanism that switches PKA from an activator to a repressor is not known. Here we show that Hh signalling activation causes PKA to switch its substrates from Ci to Smo within the Hh signalling complex (HSC). In particular, Hh signalling increases the level of Smo, which then outcompetes Ci for association with PKA and causes a switch in PKA substrate recognition. We propose a new model in which the PKA is constitutively present and active within the HSC, and in which the relative levels of Ci and Smo within the HSC determine differential activation and cellular response to Hh signalling.
Collapse
Affiliation(s)
- Nadia Ranieri
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Pascal P Thérond
- 1] University Nice Sophia Antipolis, iBV, UMR 7277, 06100 Nice, France [2] CNRS, iBV, UMR 7277, Parc Valrose, 06100 Nice, France [3] INSERM, iBV, U10891, 06100 Nice, France
| | - Laurent Ruel
- 1] University Nice Sophia Antipolis, iBV, UMR 7277, 06100 Nice, France [2] CNRS, iBV, UMR 7277, Parc Valrose, 06100 Nice, France [3] INSERM, iBV, U10891, 06100 Nice, France
| |
Collapse
|
50
|
Sonic hedgehog signalling pathway: a complex network. Ann Neurosci 2014; 21:28-31. [PMID: 25206052 PMCID: PMC4117150 DOI: 10.5214/ans.0972.7531.210109] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 01/11/2014] [Accepted: 02/25/2014] [Indexed: 12/21/2022] Open
Abstract
Sonic Hedgehog (Shh) signalling cascade is one of the intricate signal transduction mechanisms that govern the precisely regulated developmental processes of multicellular organisms. Along with establishing the patterns of cellular differentiation to direct complex organ formation, it also has an important role in post-embryonic tissue regeneration and repair processes. Especially, Shh signalling is implicated in the induction of multifarious neuronal populations in central nervous system. There is compelling evidence of the involvement of Shh protein in the signalling network that regulates various morphogenetic processes such as the exquisite neural tube pattern formation. In the morphogenetic field, the activation of Shh signalling processes is intricately linked to the alterations at the molecular level in the structure of Shh protein that leads to its altered biophysical and biochemical reactivity. This brief article gives an overview of such complex cascade of events in Shh signalling and its transduction pathways.
Collapse
|