1
|
Øye H, Lundekvam M, Caiella A, Hellesvik M, Arnesen T. Protein N-terminal modifications: molecular machineries and biological implications. Trends Biochem Sci 2025; 50:290-310. [PMID: 39837675 DOI: 10.1016/j.tibs.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/15/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025]
Abstract
The majority of eukaryotic proteins undergo N-terminal (Nt) modifications facilitated by various enzymes. These enzymes, which target the initial amino acid of a polypeptide in a sequence-dependent manner, encompass peptidases, transferases, cysteine oxygenases, and ligases. Nt modifications - such as acetylation, fatty acylations, methylation, arginylation, and oxidation - enhance proteome complexity and regulate protein targeting, stability, and complex formation. Modifications at protein N termini are thereby core components of a large number of biological processes, including cell signaling and motility, autophagy regulation, and plant and animal oxygen sensing. Dysregulation of Nt-modifying enzymes is implicated in several human diseases. In this feature review we provide an overview of the various protein Nt modifications occurring either co- or post-translationally, the enzymes involved, and the biological impact.
Collapse
Affiliation(s)
- Hanne Øye
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| | - Malin Lundekvam
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Alessia Caiella
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Thomas Arnesen
- Department of Biomedicine, University of Bergen, Bergen, Norway; Department of Surgery, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
2
|
Fan Z, Hao Y, Huo Y, Cao F, Li L, Xu J, Song Y, Yang K. Modulators for palmitoylation of proteins and small molecules. Eur J Med Chem 2024; 271:116408. [PMID: 38621327 DOI: 10.1016/j.ejmech.2024.116408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024]
Abstract
As an essential form of lipid modification for maintaining vital cellular functions, palmitoylation plays an important role in in the regulation of various physiological processes, serving as a promising therapeutic target for diseases like cancer and neurological disorders. Ongoing research has revealed that palmitoylation can be categorized into three distinct types: N-palmitoylation, O-palmitoylation and S-palmitoylation. Herein this paper provides an overview of the regulatory enzymes involved in palmitoylation, including palmitoyltransferases and depalmitoylases, and discusses the currently available broad-spectrum and selective inhibitors for these enzymes.
Collapse
Affiliation(s)
- Zeshuai Fan
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Yuchen Hao
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Yidan Huo
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China
| | - Fei Cao
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei, 071002, China
| | - Longfei Li
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei, 071002, China
| | - Jianmei Xu
- Department of hematopathology, Affiliated Hospital of Hebei University, Hebei University, Baoding, 071002, China
| | - Yali Song
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei, 071002, China
| | - Kan Yang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, College of Pharmaceutical Sciences, Hebei University, Baoding, 071002, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding, Hebei, 071002, China.
| |
Collapse
|
3
|
Ehlers SF, Manikowski D, Steffes G, Ehring K, Gude F, Grobe K. A Residual N-Terminal Peptide Enhances Signaling of Depalmitoylated Hedgehog to the Patched Receptor. J Dev Biol 2024; 12:11. [PMID: 38651456 PMCID: PMC11036296 DOI: 10.3390/jdb12020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
During their biosynthesis, Sonic hedgehog (Shh) morphogens are covalently modified by cholesterol at the C-terminus and palmitate at the N-terminus. Although both lipids initially anchor Shh to the plasma membrane of producing cells, it later translocates to the extracellular compartment to direct developmental fates in cells expressing the Patched (Ptch) receptor. Possible release mechanisms for dually lipidated Hh/Shh into the extracellular compartment are currently under intense debate. In this paper, we describe the serum-dependent conversion of the dually lipidated cellular precursor into a soluble cholesteroylated variant (ShhC) during its release. Although ShhC is formed in a Dispatched- and Scube2-dependent manner, suggesting the physiological relevance of the protein, the depalmitoylation of ShhC during release is inconsistent with the previously postulated function of N-palmitate in Ptch receptor binding and signaling. Therefore, we analyzed the potency of ShhC to induce Ptch-controlled target cell transcription and differentiation in Hh-sensitive reporter cells and in the Drosophila eye. In both experimental systems, we found that ShhC was highly bioactive despite the absence of the N-palmitate. We also found that the artificial removal of N-terminal peptides longer than eight amino acids inactivated the depalmitoylated soluble proteins in vitro and in the developing Drosophila eye. These results demonstrate that N-depalmitoylated ShhC requires an N-peptide of a defined minimum length for its signaling function to Ptch.
Collapse
Affiliation(s)
- Sophia F. Ehlers
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Georg Steffes
- Institute for Neuro- and Behavioral Biology, Faculty of Biology, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany;
| | - Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, Faculty of Medicine, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany; (S.F.E.); (D.M.); (K.E.); (F.G.)
| |
Collapse
|
4
|
Ritzefeld M, Zhang L, Xiao Z, Andrei SA, Boyd O, Masumoto N, Rodgers UR, Artelsmair M, Sefer L, Hayes A, Gavriil ES, Raynaud FI, Burke R, Blagg J, Rzepa HS, Siebold C, Magee AI, Lanyon-Hogg T, Tate EW. Design, Synthesis, and Evaluation of Inhibitors of Hedgehog Acyltransferase. J Med Chem 2024; 67:1061-1078. [PMID: 38198226 PMCID: PMC10823475 DOI: 10.1021/acs.jmedchem.3c01363] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/08/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024]
Abstract
Hedgehog signaling is involved in embryonic development and cancer growth. Functional activity of secreted Hedgehog signaling proteins is dependent on N-terminal palmitoylation, making the palmitoyl transferase Hedgehog acyltransferase (HHAT), a potential drug target and a series of 4,5,6,7-tetrahydrothieno[3,2-c]pyridines have been identified as HHAT inhibitors. Based on structural data, we designed and synthesized 37 new analogues which we profiled alongside 13 previously reported analogues in enzymatic and cellular assays. Our results show that a central amide linkage, a secondary amine, and (R)-configuration at the 4-position of the core are three key factors for inhibitory potency. Several potent analogues with low- or sub-μM IC50 against purified HHAT also inhibit Sonic Hedgehog (SHH) palmitoylation in cells and suppress the SHH signaling pathway. This work identifies IMP-1575 as the most potent cell-active chemical probe for HHAT function, alongside an inactive control enantiomer, providing tool compounds for validation of HHAT as a target in cellular assays.
Collapse
Affiliation(s)
- Markus Ritzefeld
- Department
of Chemistry, Imperial College London, London W12 0BZ, U.K.
| | - Leran Zhang
- Department
of Chemistry, Imperial College London, London W12 0BZ, U.K.
| | - Zhangping Xiao
- Department
of Chemistry, Imperial College London, London W12 0BZ, U.K.
| | | | - Olivia Boyd
- Department
of Chemistry, Imperial College London, London W12 0BZ, U.K.
| | - Naoko Masumoto
- Department
of Chemistry, Imperial College London, London W12 0BZ, U.K.
| | - Ursula R. Rodgers
- National
Heart and Lung Institute, Imperial College
London, London SW7 2AZ, U.K.
| | - Markus Artelsmair
- Department
of Chemistry, Imperial College London, London W12 0BZ, U.K.
| | - Lea Sefer
- Division
of Structural Biology, University of Oxford, Oxford OX3 7BN, U.K.
| | - Angela Hayes
- Division
of Cancer Therapeutics, Centre for Cancer Drug Discovery, Institute of Cancer Research, London SM2 5NG, U.K.
| | | | - Florence I. Raynaud
- Division
of Cancer Therapeutics, Centre for Cancer Drug Discovery, Institute of Cancer Research, London SM2 5NG, U.K.
| | - Rosemary Burke
- Division
of Cancer Therapeutics, Centre for Cancer Drug Discovery, Institute of Cancer Research, London SM2 5NG, U.K.
| | - Julian Blagg
- Division
of Cancer Therapeutics, Centre for Cancer Drug Discovery, Institute of Cancer Research, London SM2 5NG, U.K.
| | - Henry S. Rzepa
- Department
of Chemistry, Imperial College London, London W12 0BZ, U.K.
| | - Christian Siebold
- Division
of Structural Biology, University of Oxford, Oxford OX3 7BN, U.K.
| | - Anthony I. Magee
- National
Heart and Lung Institute, Imperial College
London, London SW7 2AZ, U.K.
| | | | - Edward W. Tate
- Department
of Chemistry, Imperial College London, London W12 0BZ, U.K.
| |
Collapse
|
5
|
Coupland CE, Ansell TB, Sansom MSP, Siebold C. Rocking the MBOAT: Structural insights into the membrane bound O-acyltransferase family. Curr Opin Struct Biol 2023; 80:102589. [PMID: 37040671 DOI: 10.1016/j.sbi.2023.102589] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 04/13/2023]
Abstract
The membrane-bound O-acyltransferase (MBOAT) superfamily catalyses the transfer of acyl chains to substrates implicated in essential cellular functions. Aberrant function of MBOATs is associated with various diseases and MBOATs are promising drug targets. There has been recent progress in structural characterisation of MBOATs, advancing our understanding of their functional mechanism. Integrating information across the MBOAT family, we characterise a common MBOAT fold and provide a blueprint for substrate and inhibitor engagement. This work provides context for the diverse substrates, mechanisms, and evolutionary relationships of protein and small-molecule MBOATs. Further work should aim to characterise MBOATs, as inherently lipid-associated proteins, within their membrane environment.
Collapse
Affiliation(s)
- Claire E Coupland
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - T Bertie Ansell
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.
| |
Collapse
|
6
|
Rjiba K, Mougou-Zerelli S, Hamida IH, Saad G, Khadija B, Jelloul A, Slimani W, Hasni Y, Dimassi S, Khelifa HB, Sallem A, Kammoun M, Abdallah HH, Gribaa M, Bignon-Topalovic J, Chelly S, Khairi H, Bibi M, Kacem M, Saad A, Bashamboo A, McElreavey K. Additional evidence for the role of chromosomal imbalances and SOX8, ZNRF3 and HHAT gene variants in early human testis development. Reprod Biol Endocrinol 2023; 21:2. [PMID: 36631813 PMCID: PMC9990451 DOI: 10.1186/s12958-022-01045-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/01/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Forty-six ,XY Differences/Disorders of Sex Development (DSD) are characterized by a broad phenotypic spectrum ranging from typical female to male with undervirilized external genitalia, or more rarely testicular regression with a typical male phenotype. Despite progress in the genetic diagnosis of DSD, most 46,XY DSD cases remain idiopathic. METHODS To determine the genetic causes of 46,XY DSD, we studied 165 patients of Tunisian ancestry, who presented a wide range of DSD phenotypes. Karyotyping, candidate gene sequencing, and whole-exome sequencing (WES) were performed. RESULTS Cytogenetic abnormalities, including a high frequency of sex chromosomal anomalies (85.4%), explained the phenotype in 30.9% (51/165) of the cohort. Sanger sequencing of candidate genes identified a novel pathogenic variant in the SRY gene in a patient with 46,XY gonadal dysgenesis. An exome screen of a sub-group of 44 patients with 46,XY DSD revealed pathogenic or likely pathogenic variants in 38.6% (17/44) of patients. CONCLUSION Rare or novel pathogenic variants were identified in the AR, SRD5A2, ZNRF3, SOX8, SOX9 and HHAT genes. Overall our data indicate a genetic diagnosis rate of 41.2% (68/165) in the group of 46,XY DSD.
Collapse
Affiliation(s)
- Khouloud Rjiba
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
- Higher Institute of Biotechnology Monastir, University of Monastir, Monastir, Tunisia
- Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
- Human Developmental Genetics Unit, CNRS UMR 3738, Institut Pasteur, Paris, France
| | - Soumaya Mougou-Zerelli
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
- Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Imen Hadj Hamida
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Ghada Saad
- Department of Endocrinology, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Bochra Khadija
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
- Higher Institute of Biotechnology Monastir, University of Monastir, Monastir, Tunisia
- Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Afef Jelloul
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Wafa Slimani
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
- Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Yosra Hasni
- Department of Endocrinology, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Sarra Dimassi
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
- Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Hela Ben Khelifa
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Amira Sallem
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
- Laboratory of Human Cytogenetics and Biology of Reproduction, Fattouma Bourguiba University Teaching Hospital, Monastir, Tunisia
| | - Molka Kammoun
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Hamza Hadj Abdallah
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Moez Gribaa
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | | | - Sami Chelly
- Private Gynecologist Sousse, Sousse, Tunisia
| | - Hédi Khairi
- Department of Gynecology and Obstetrics, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Mohamed Bibi
- Department of Gynecology and Obstetrics, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Maha Kacem
- Department of Endocrinology, Farhat Hached University Teaching Hospital, Sousse, Tunisia
| | - Ali Saad
- Laboratory of Human Cytogenetics, Molecular Genetics and Biology of Human Reproduction, Farhat Hached University Teaching Hospital, Sousse, Tunisia
- Unité de Services Communs en Génétique Humaine, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Anu Bashamboo
- Human Developmental Genetics Unit, CNRS UMR 3738, Institut Pasteur, Paris, France
| | - Kenneth McElreavey
- Human Developmental Genetics Unit, CNRS UMR 3738, Institut Pasteur, Paris, France.
| |
Collapse
|
7
|
Nguyen PK, Cheng LY. Non-autonomous regulation of neurogenesis by extrinsic cues: a Drosophila perspective. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac004. [PMID: 38596708 PMCID: PMC10913833 DOI: 10.1093/oons/kvac004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 04/11/2024]
Abstract
The formation of a functional circuitry in the central nervous system (CNS) requires the correct number and subtypes of neural cells. In the developing brain, neural stem cells (NSCs) self-renew while giving rise to progenitors that in turn generate differentiated progeny. As such, the size and the diversity of cells that make up the functional CNS depend on the proliferative properties of NSCs. In the fruit fly Drosophila, where the process of neurogenesis has been extensively investigated, extrinsic factors such as the microenvironment of NSCs, nutrients, oxygen levels and systemic signals have been identified as regulators of NSC proliferation. Here, we review decades of work that explores how extrinsic signals non-autonomously regulate key NSC characteristics such as quiescence, proliferation and termination in the fly.
Collapse
Affiliation(s)
- Phuong-Khanh Nguyen
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Victoria 3010, Australia
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
8
|
Coupland CE, Andrei SA, Ansell TB, Carrique L, Kumar P, Sefer L, Schwab RA, Byrne EFX, Pardon E, Steyaert J, Magee AI, Lanyon-Hogg T, Sansom MSP, Tate EW, Siebold C. Structure, mechanism, and inhibition of Hedgehog acyltransferase. Mol Cell 2021; 81:5025-5038.e10. [PMID: 34890564 PMCID: PMC8693861 DOI: 10.1016/j.molcel.2021.11.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/27/2021] [Accepted: 11/17/2021] [Indexed: 01/20/2023]
Abstract
The Sonic Hedgehog (SHH) morphogen pathway is fundamental for embryonic development and stem cell maintenance and is implicated in various cancers. A key step in signaling is transfer of a palmitate group to the SHH N terminus, catalyzed by the multi-pass transmembrane enzyme Hedgehog acyltransferase (HHAT). We present the high-resolution cryo-EM structure of HHAT bound to substrate analog palmityl-coenzyme A and a SHH-mimetic megabody, revealing a heme group bound to HHAT that is essential for HHAT function. A structure of HHAT bound to potent small-molecule inhibitor IMP-1575 revealed conformational changes in the active site that occlude substrate binding. Our multidisciplinary analysis provides a detailed view of the mechanism by which HHAT adapts the membrane environment to transfer an acyl chain across the endoplasmic reticulum membrane. This structure of a membrane-bound O-acyltransferase (MBOAT) superfamily member provides a blueprint for other protein-substrate MBOATs and a template for future drug discovery.
Collapse
Affiliation(s)
- Claire E Coupland
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Sebastian A Andrei
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - T Bertie Ansell
- Department of Biochemistry, University of Oxford, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Loic Carrique
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Pramod Kumar
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Lea Sefer
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Rebekka A Schwab
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Eamon F X Byrne
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Els Pardon
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050 Brussels, Belgium; VIB-VUB Center for Structural Biology, Vlaams Instituut Biotechnologie (VIB), Pleinlaan 2, 1050 Brussels, Belgium
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, 1050 Brussels, Belgium; VIB-VUB Center for Structural Biology, Vlaams Instituut Biotechnologie (VIB), Pleinlaan 2, 1050 Brussels, Belgium
| | - Anthony I Magee
- National Heart and Lung Institute, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Thomas Lanyon-Hogg
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, 82 Wood Lane, London W12 0BZ, UK.
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK.
| |
Collapse
|
9
|
Ehring K, Manikowski D, Goretzko J, Froese J, Gude F, Jakobs P, Rescher U, Kirchhefer U, Grobe K. Conserved cholesterol-related activities of Dispatched 1 drive Sonic hedgehog shedding from the cell membrane. J Cell Sci 2021; 135:271842. [PMID: 34308968 PMCID: PMC8403983 DOI: 10.1242/jcs.258672] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/08/2021] [Indexed: 01/04/2023] Open
Abstract
The Sonic hedgehog (Shh) pathway controls embryonic development and tissue homeostasis after birth. Long-standing questions about this pathway include how the dual-lipidated, firmly plasma membrane-associated Shh ligand is released from producing cells to signal to distant target cells and how the resistance-nodulation-division transporter Dispatched 1 (Disp, also known as Disp1) regulates this process. Here, we show that inactivation of Disp in Shh-expressing human cells impairs proteolytic Shh release from its lipidated terminal peptides, a process called ectodomain shedding. We also show that cholesterol export from Disp-deficient cells is reduced, that these cells contain increased cholesterol amounts in the plasma membrane, and that Shh shedding from Disp-deficient cells is restored by pharmacological membrane cholesterol extraction and by overexpression of transgenic Disp or the structurally related protein Patched 1 (Ptc, also known as Ptch1; a putative cholesterol transporter). These data suggest that Disp can regulate Shh function via controlled cell surface shedding and that membrane cholesterol-related molecular mechanisms shared by Disp and Ptc exercise such sheddase control.
Collapse
Affiliation(s)
- Kristina Ehring
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Jonas Goretzko
- Center for Molecular Biology of Inflammation, Institute for Medical Biochemistry, University of Münster, Von Esmarch Strasse 56, D-48149 Münster, Germany
| | - Jurij Froese
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Fabian Gude
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | - Ursula Rescher
- Center for Molecular Biology of Inflammation, Institute for Medical Biochemistry, University of Münster, Von Esmarch Strasse 56, D-48149 Münster, Germany
| | - Uwe Kirchhefer
- Institute of Pharmacology and Toxicology, University of Münster, Domagkstrasse 12, D-48149 Münster, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| |
Collapse
|
10
|
Jiang Y, Benz TL, Long SB. Substrate and product complexes reveal mechanisms of Hedgehog acylation by HHAT. Science 2021; 372:1215-1219. [PMID: 34112694 DOI: 10.1126/science.abg4998] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 05/13/2021] [Indexed: 12/22/2022]
Abstract
Hedgehog proteins govern crucial developmental steps in animals and drive certain human cancers. Before they can function as signaling molecules, Hedgehog precursor proteins must undergo amino-terminal palmitoylation by Hedgehog acyltransferase (HHAT). We present cryo-electron microscopy structures of human HHAT in complex with its palmitoyl-coenzyme A substrate and of a product complex with a palmitoylated Hedgehog peptide at resolutions of 2.7 and 3.2 angstroms, respectively. The structures reveal how HHAT overcomes the challenges of bringing together substrates that have different physiochemical properties from opposite sides of the endoplasmic reticulum membrane within a membrane-embedded active site for catalysis. These principles are relevant to related enzymes that catalyze the acylation of Wnt and of the appetite-stimulating hormone ghrelin. The structural and mechanistic insights may advance the development of inhibitors for cancer.
Collapse
Affiliation(s)
- Yiyang Jiang
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Thomas L Benz
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen B Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
11
|
Resh MD. Palmitoylation of Hedgehog proteins by Hedgehog acyltransferase: roles in signalling and disease. Open Biol 2021; 11:200414. [PMID: 33653085 PMCID: PMC8061759 DOI: 10.1098/rsob.200414] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hedgehog acyltransferase (Hhat), a member of the membrane-bound O-acyltransferase (MBOAT) family, catalyses the covalent attachment of palmitate to the N-terminus of Hedgehog proteins. Palmitoylation is a post-translational modification essential for Hedgehog signalling. This review explores the mechanisms involved in Hhat acyltransferase enzymatic activity, similarities and differences between Hhat and other MBOAT enzymes, and the role of palmitoylation in Hedgehog signalling. In vitro and cell-based assays for Hhat activity have been developed, and residues within Hhat and Hedgehog essential for palmitoylation have been identified. In cells, Hhat promotes the transfer of palmitoyl-CoA from the cytoplasmic to the luminal side of the endoplasmic reticulum membrane, where Shh palmitoylation occurs. Palmitoylation is required for efficient delivery of secreted Hedgehog to its receptor Patched1, as well as for the deactivation of Patched1, which initiates the downstream Hedgehog signalling pathway. While Hhat loss is lethal during embryogenesis, mutations in Hhat have been linked to disease states or abnormalities in mice and humans. In adults, aberrant re-expression of Hedgehog ligands promotes tumorigenesis in an Hhat-dependent manner in a variety of different cancers, including pancreatic, breast and lung. Targeting hedgehog palmitoylation by inhibition of Hhat is thus a promising, potential intervention in human disease.
Collapse
Affiliation(s)
- Marilyn D Resh
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 143, New York, NY 10065, USA
| |
Collapse
|
12
|
Hong S, Hu P, Jang JH, Carrington B, Sood R, Berger SI, Roessler E, Muenke M. Functional analysis of Sonic Hedgehog variants associated with holoprosencephaly in humans using a CRISPR/Cas9 zebrafish model. Hum Mutat 2020; 41:2155-2166. [PMID: 32939873 DOI: 10.1002/humu.24119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/17/2020] [Accepted: 09/12/2020] [Indexed: 01/20/2023]
Abstract
Genetic variation in the highly conserved Sonic Hedgehog (SHH) gene is one of the most common genetic causes for the malformations of the brain and face in humans described as the holoprosencephaly clinical spectrum. However, only a minor fraction of known SHH variants have been experimentally proven to lead to abnormal function. Employing a phenotypic rescue assay with synthetic human messenger RNA variant constructs in shha-/- knockout zebrafish, we evaluated 104 clinically reported in-frame and missense SHH variants. Our data helped us to classify them into loss of function variants (31), hypomorphic variants (33), and nonpathogenic variants (40). We discuss the strengths and weaknesses of currently accepted predictors of variant deleteriousness and the American College of Medical Genetics and Genomics guidelines for variant interpretation in the context of this functional model; furthermore, we demonstrate the robustness of model systems such as zebrafish as a rapid method to resolve variants of uncertain significance.
Collapse
Affiliation(s)
- Sungkook Hong
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Hu
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jae Hee Jang
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA.,College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, Maryland, USA
| | - Blake Carrington
- Zebrafish Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Raman Sood
- Zebrafish Core, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Seth I Berger
- Children's National Hospital, Center for Genetic Medicine Research and Rare Disease Institute, Washington DC, USA
| | - Erich Roessler
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Maximilian Muenke
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA.,American College of Medical Genetics and Genomics, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Sasai N, Toriyama M, Kondo T. Hedgehog Signal and Genetic Disorders. Front Genet 2019; 10:1103. [PMID: 31781166 PMCID: PMC6856222 DOI: 10.3389/fgene.2019.01103] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
The hedgehog (Hh) family comprises sonic hedgehog (Shh), Indian hedgehog (Ihh), and desert hedgehog (Dhh), which are versatile signaling molecules involved in a wide spectrum of biological events including cell differentiation, proliferation, and survival; establishment of the vertebrate body plan; and aging. These molecules play critical roles from embryogenesis to adult stages; therefore, alterations such as abnormal expression or mutations of the genes involved and their downstream factors cause a variety of genetic disorders at different stages. The Hh family involves many signaling mediators and functions through complex mechanisms, and achieving a comprehensive understanding of the entire signaling system is challenging. This review discusses the signaling mediators of the Hh pathway and their functions at the cellular and organismal levels. We first focus on the roles of Hh signaling mediators in signal transduction at the cellular level and the networks formed by these factors. Then, we analyze the spatiotemporal pattern of expression of Hh pathway molecules in tissues and organs, and describe the phenotypes of mutant mice. Finally, we discuss the genetic disorders caused by malfunction of Hh signaling-related molecules in humans.
Collapse
Affiliation(s)
- Noriaki Sasai
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Michinori Toriyama
- Systems Neurobiology and Medicine, Division of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
14
|
Lanyon-Hogg T, Ritzefeld M, Sefer L, Bickel JK, Rudolf AF, Panyain N, Bineva-Todd G, Ocasio CA, O'Reilly N, Siebold C, Magee AI, Tate EW. Acylation-coupled lipophilic induction of polarisation (Acyl-cLIP): a universal assay for lipid transferase and hydrolase enzymes. Chem Sci 2019; 10:8995-9000. [PMID: 31762980 PMCID: PMC6855259 DOI: 10.1039/c9sc01785b] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022] Open
Abstract
Posttranslational attachment of lipids to proteins is important for many cellular functions, and the enzymes responsible for these modifications are implicated in many diseases, from cancer to neurodegeneration. Lipid transferases and hydrolases are increasingly tractable therapeutic targets, but present unique challenges for high-throughput biochemical enzyme assays which hinder development of new inhibitors. We present Acylation-coupled Lipophilic Induction of Polarisation (Acyl-cLIP) as the first universally applicable biochemical lipidation assay, exploiting the hydrophobic nature of lipidated peptides to drive a polarised fluorescence readout. Acyl-cLIP allows sensitive, accurate, real-time measurement of S- or N-palmitoylation, N-myristoylation, S-farnesylation or S-geranylgeranylation. Furthermore, it is applicable to transfer and hydrolysis reactions, and we demonstrate its extension to a high-throughput screening format. We anticipate that Acyl-cLIP will greatly expedite future drug discovery efforts against these challenging targets.
Collapse
Affiliation(s)
- Thomas Lanyon-Hogg
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| | - Markus Ritzefeld
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| | - Lea Sefer
- Division of Structural Biology , Wellcome Centre for Human Genetics , University of Oxford , Oxford , OX3 7BN , UK
| | - Jasmine K Bickel
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| | - Amalie F Rudolf
- Division of Structural Biology , Wellcome Centre for Human Genetics , University of Oxford , Oxford , OX3 7BN , UK
| | - Nattawadee Panyain
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| | | | | | | | - Christian Siebold
- Division of Structural Biology , Wellcome Centre for Human Genetics , University of Oxford , Oxford , OX3 7BN , UK
| | - Anthony I Magee
- Molecular Medicine Section , National Heart & Lung Institute , Imperial College London , London , SW7 2AZ , UK
| | - Edward W Tate
- Department of Chemistry , Imperial College London , London , W12 0BZ , UK . ; ; Tel: +44 0207 5943752 ; Tel: +44 0207 5945821
| |
Collapse
|
15
|
Hu A, Song BL. The interplay of Patched, Smoothened and cholesterol in Hedgehog signaling. Curr Opin Cell Biol 2019; 61:31-38. [PMID: 31369952 DOI: 10.1016/j.ceb.2019.06.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/29/2019] [Accepted: 06/30/2019] [Indexed: 12/29/2022]
Abstract
The Hedgehog (HH) pathway plays a pivotal role in regulating a diverse array of events from embryonic tissue patterning to adult organ self-renewal. Aberrant activation of the pathway is linked to carcinogenesis. Key factors in the HH pathway include the signaling ligand HH, the receptor Patched (PTCH), and the G-protein-coupled receptor-like transducer Smoothened (SMO). A long-lasting question about this pathway is how PTCH prevents SMO from being activated. Recent high-resolution structural studies provide insight into the molecular basis of HH recognition by PTCH. Moreover, cholesterol stands out as the endogenous ligand of SMO and acts by binding and/or covalently linking to SMO. In this review, we discuss current advances in HH signaling, the interplay of PTCH, SMO and cholesterol, and propose putative models of SMO activation by cholesterol binding and/or modification.
Collapse
Affiliation(s)
- Ao Hu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
16
|
Schürmann S, Steffes G, Manikowski D, Kastl P, Malkus U, Bandari S, Ohlig S, Ortmann C, Rebollido-Rios R, Otto M, Nüsse H, Hoffmann D, Klämbt C, Galic M, Klingauf J, Grobe K. Proteolytic processing of palmitoylated Hedgehog peptides specifies the 3-4 intervein region of the Drosophila wing. eLife 2018. [PMID: 29522397 PMCID: PMC5844694 DOI: 10.7554/elife.33033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cell fate determination during development often requires morphogen transport from producing to distant responding cells. Hedgehog (Hh) morphogens present a challenge to this concept, as all Hhs are synthesized as terminally lipidated molecules that form insoluble clusters at the surface of producing cells. While several proposed Hh transport modes tie directly into these unusual properties, the crucial step of Hh relay from producing cells to receptors on remote responding cells remains unresolved. Using wing development in Drosophila melanogaster as a model, we show that Hh relay and direct patterning of the 3–4 intervein region strictly depend on proteolytic removal of lipidated N-terminal membrane anchors. Site-directed modification of the N-terminal Hh processing site selectively eliminated the entire 3–4 intervein region, and additional targeted removal of N-palmitate restored its formation. Hence, palmitoylated membrane anchors restrict morphogen spread until site-specific processing switches membrane-bound Hh into bioactive forms with specific patterning functions. Each cell in a developing embryo receives information that determines what type of body structure it will form. In fruit flies, this information is partly given by a protein called Hedgehog. In the embryo cells that receive it, Hedgehog can trigger a series of events which activate certain genes and thereby regulate structure formation. The Hedgehog proteins are produced by a different organizing group of cells: from there they transport within the embryo, creating a gradient. Depending on where a responding cell is in the embryo, it receives a different amount of Hedgehog, which gives the cell its identity. For example, Hedgehog proteins form a gradient across a fruit fly’s developing wing, which creates a visible vein pattern. How Hedgehog proteins form gradients is enigmatic, however, because once produced, they cling to the cells that created them. The reason for this unusual behavior is that the two ends of the Hedgehog protein are attached to a different fat molecule. In particular, one extremity is linked to a fat molecule called palmitate. These ends’ fatty additions anchor Hedgehog to the cells that produced them. Then, the tethered proteins gather together to form chain-like clusters where they inactivate each other: the extremity with the palmitate ‘hides’ the portion of the neighboring protein that binds to the receiving cells. It is still unclear how Hedgehog can be activated and released to reach these faraway cells. One hypothesis is that an enzyme comes to the clusters and frees the proteins by cutting both of Hedgehog’s fatty anchors. Thanks to how the palmitate tethers Hedgehog to the cell, the protein is positioned in such a way that when the enzyme makes its snip, the binding site on the neighboring Hedgehog gets exposed: this protein is activated and, when also cut by the enzyme, released. Here, Schürmann et al. create an array of mutant Hedgehog proteins – for example some without palmitate, some with palmitate that cannot be removed by the enzyme – and study how they affect the development of the wing’s pattern in the fruit fly. Coupled with the imaging of the clusters, these experiments support the hypothesis that the palmitate anchor is necessary so that Hedgehog proteins can be turned on before diffusing away. The Hedgehog family of proteins is also present in humans, where it presides over the development of the embryo but is also involved in cancer. Understanding how Hedgehog works in the fruit fly could lead to new discoveries in humans too.
Collapse
Affiliation(s)
- Sabine Schürmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Georg Steffes
- Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany.,Institute of Neurobiology, University of Münster, Münster, Germany
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Philipp Kastl
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Ursula Malkus
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Shyam Bandari
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Stefanie Ohlig
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Corinna Ortmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | | | - Mandy Otto
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| | - Harald Nüsse
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Daniel Hoffmann
- Center for Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Christian Klämbt
- Institute of Neurobiology, University of Münster, Münster, Germany
| | - Milos Galic
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Münster, Germany
| |
Collapse
|
17
|
Manikowski D, Kastl P, Grobe K. Taking the Occam's Razor Approach to Hedgehog Lipidation and Its Role in Development. J Dev Biol 2018; 6:jdb6010003. [PMID: 29615552 PMCID: PMC5875562 DOI: 10.3390/jdb6010003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 01/01/2023] Open
Abstract
All Hedgehog (Hh) proteins signal from producing cells to distant receiving cells despite being synthesized as N-and C-terminally lipidated, membrane-tethered molecules. To explain this paradoxical situation, over the past 15 years, several hypotheses have been postulated that tie directly into this property, such as Hh transport on cellular extensions called cytonemes or on secreted vesicles called lipophorins and exosomes. The alternative situation that tight membrane association merely serves to prevent unregulated Hh solubilization has been addressed by biochemical and structural studies suggesting Hh extraction from the membrane or proteolytic Hh release. While some of these models may act in different organisms, tissues or developmental programs, others may act together to specify Hh short- and long-range signaling in the same tissues. To test and rank these possibilities, we here review major models of Hh release and transport and hypothesize that the (bio)chemical and physical properties of firmly established, homologous, and functionally essential biochemical Hh modifications are adapted to specify and determine interdependent steps of Hh release, transport and signaling, while ruling out other steps. This is also described by the term “congruence”, meaning that the logical combination of biochemical Hh modifications can reveal their true functional implications. This combined approach reveals potential links between models of Hh release and transport that were previously regarded as unrelated, thereby expanding our view of how Hhs can steer development in a simple, yet extremely versatile, manner.
Collapse
Affiliation(s)
- Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| | - Philipp Kastl
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence, University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
18
|
Jiang H, Zhang X, Chen X, Aramsangtienchai P, Tong Z, Lin H. Protein Lipidation: Occurrence, Mechanisms, Biological Functions, and Enabling Technologies. Chem Rev 2018; 118:919-988. [PMID: 29292991 DOI: 10.1021/acs.chemrev.6b00750] [Citation(s) in RCA: 331] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein lipidation, including cysteine prenylation, N-terminal glycine myristoylation, cysteine palmitoylation, and serine and lysine fatty acylation, occurs in many proteins in eukaryotic cells and regulates numerous biological pathways, such as membrane trafficking, protein secretion, signal transduction, and apoptosis. We provide a comprehensive review of protein lipidation, including descriptions of proteins known to be modified and the functions of the modifications, the enzymes that control them, and the tools and technologies developed to study them. We also highlight key questions about protein lipidation that remain to be answered, the challenges associated with answering such questions, and possible solutions to overcome these challenges.
Collapse
Affiliation(s)
- Hong Jiang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University , Ithaca, New York 14853, United States
| | - Xiaoyu Zhang
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University , Ithaca, New York 14853, United States
| | - Xiao Chen
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University , Ithaca, New York 14853, United States
| | - Pornpun Aramsangtienchai
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University , Ithaca, New York 14853, United States
| | - Zhen Tong
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University , Ithaca, New York 14853, United States
| | - Hening Lin
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University , Ithaca, New York 14853, United States
| |
Collapse
|
19
|
Jakobs P, Schulz P, Schürmann S, Niland S, Exner S, Rebollido-Rios R, Manikowski D, Hoffmann D, Seidler DG, Grobe K. Ca 2+ coordination controls sonic hedgehog structure and its Scube2-regulated release. J Cell Sci 2017; 130:3261-3271. [PMID: 28778988 DOI: 10.1242/jcs.205872] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/30/2017] [Indexed: 12/22/2022] Open
Abstract
Proteolytic processing of cell-surface-bound ligands, called shedding, is a fundamental system to control cell-cell signaling. Yet, our understanding of how shedding is regulated is still incomplete. One way to increase the processing of dual-lipidated membrane-associated Sonic hedgehog (Shh) is to increase the density of substrate and sheddase. This releases and also activates Shh by the removal of lipidated inhibitory N-terminal peptides from Shh receptor binding sites. Shh release and activation is enhanced by Scube2 [signal sequence, cubulin (CUB) domain, epidermal growth factor (EGF)-like protein 2], raising the question of how this is achieved. Here, we show that Scube2 EGF domains are responsible for specific proteolysis of the inhibitory Shh N-terminus, and that CUB domains complete the process by reversing steric masking of this peptide. Steric masking, in turn, depends on Ca2+ occupancy of Shh ectodomains, unveiling a new mode of shedding regulation at the substrate level. Importantly, Scube2 uncouples processing of Shh peptides from their lipid-mediated juxtamembrane positioning, and thereby explains the long-standing conundrum that N-terminally unlipidated Shh shows patterning activity in Scube2-expressing vertebrates, but not in invertebrates that lack Scube orthologs.
Collapse
Affiliation(s)
- Petra Jakobs
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Philipp Schulz
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Sabine Schürmann
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Sebastian Exner
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Rocio Rebollido-Rios
- Center for Medical Biotechnology, University of Duisburg-Essen, D-45117 Essen, Germany
| | - Dominique Manikowski
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| | - Daniel Hoffmann
- Center for Medical Biotechnology, University of Duisburg-Essen, D-45117 Essen, Germany
| | - Daniela G Seidler
- Centre for Internal Medicine, Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School I3, EB2/R3110, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, D-48149 Münster, Germany
| |
Collapse
|
20
|
Rodgers U, Lanyon-Hogg T, Masumoto N, Ritzefeld M, Burke R, Blagg J, Magee AI, Tate EW. Characterization of Hedgehog Acyltransferase Inhibitors Identifies a Small Molecule Probe for Hedgehog Signaling by Cancer Cells. ACS Chem Biol 2016; 11:3256-3262. [PMID: 27779865 PMCID: PMC5349656 DOI: 10.1021/acschembio.6b00896] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 01/01/2023]
Abstract
The Sonic Hedgehog (Shh) signaling pathway plays a critical role during embryonic development and cancer progression. N-terminal palmitoylation of Shh by Hedgehog acyltransferase (Hhat) is essential for efficient signaling, raising interest in Hhat as a novel drug target. A recently identified series of dihydrothienopyridines has been proposed to function via this mode of action; however, the lead compound in this series (RUSKI-43) was subsequently shown to possess cytotoxic activity unrelated to canonical Shh signaling. To identify a selective chemical probe for cellular studies, we profiled three RUSKI compounds in orthogonal cell-based assays. We found that RUSKI-43 exhibits off-target cytotoxicity, masking its effect on Hhat-dependent signaling, hence results obtained with this compound in cells should be treated with caution. In contrast, RUSKI-201 showed no off-target cytotoxicity, and quantitative whole-proteome palmitoylation profiling with a bioorthogonal alkyne-palmitate reporter demonstrated specific inhibition of Hhat in cells. RUSKI-201 is the first selective Hhat chemical probe in cells and should be used in future studies of Hhat catalytic function.
Collapse
Affiliation(s)
- Ursula
R. Rodgers
- Molecular
Medicine Section, National Heart & Lung
Institute, Imperial College London, London SW7 2AZ, United
Kingdom
| | - Thomas Lanyon-Hogg
- Department
of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| | - Naoko Masumoto
- Department
of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| | - Markus Ritzefeld
- Department
of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| | - Rosemary Burke
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London, SW7 3RP, United Kingdom
| | - Julian Blagg
- Cancer
Research UK Cancer Therapeutics Unit, The
Institute of Cancer Research, London, SW7 3RP, United Kingdom
| | - Anthony I. Magee
- Molecular
Medicine Section, National Heart & Lung
Institute, Imperial College London, London SW7 2AZ, United
Kingdom
| | - Edward W. Tate
- Department
of Chemistry, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
21
|
Sena E, Feistel K, Durand BC. An Evolutionarily Conserved Network Mediates Development of the zona limitans intrathalamica, a Sonic Hedgehog-Secreting Caudal Forebrain Signaling Center. J Dev Biol 2016; 4:jdb4040031. [PMID: 29615594 PMCID: PMC5831802 DOI: 10.3390/jdb4040031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 09/29/2016] [Accepted: 10/13/2016] [Indexed: 12/16/2022] Open
Abstract
Recent studies revealed new insights into the development of a unique caudal forebrain-signaling center: the zona limitans intrathalamica (zli). The zli is the last brain signaling center to form and the first forebrain compartment to be established. It is the only part of the dorsal neural tube expressing the morphogen Sonic Hedgehog (Shh) whose activity participates in the survival, growth and patterning of neuronal progenitor subpopulations within the thalamic complex. Here, we review the gene regulatory network of transcription factors and cis-regulatory elements that underlies formation of a shh-expressing delimitated domain in the anterior brain. We discuss evidence that this network predates the origin of chordates. We highlight the contribution of Shh, Wnt and Notch signaling to zli development and discuss implications for the fact that the morphogen Shh relies on primary cilia for signal transduction. The network that underlies zli development also contributes to thalamus induction, and to its patterning once the zli has been set up. We present an overview of the brain malformations possibly associated with developmental defects in this gene regulatory network (GRN).
Collapse
Affiliation(s)
- Elena Sena
- Institut Curie, Université Paris Sud, INSERM U1021, CNRS UMR3347, Centre Universitaire, Bâtiment 110, F-91405 Orsay Cedex, France.
| | - Kerstin Feistel
- Institute of Zoology, University of Hohenheim, Garbenstr. 30, 70593 Stuttgart, Germany.
| | - Béatrice C Durand
- Institut Curie, Université Paris Sud, INSERM U1021, CNRS UMR3347, Centre Universitaire, Bâtiment 110, F-91405 Orsay Cedex, France.
| |
Collapse
|
22
|
Mechanism of inhibition of the tumor suppressor Patched by Sonic Hedgehog. Proc Natl Acad Sci U S A 2016; 113:E5866-E5875. [PMID: 27647915 DOI: 10.1073/pnas.1606719113] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Hedgehog cell-cell signaling pathway is crucial for animal development, and its misregulation is implicated in numerous birth defects and cancers. In unstimulated cells, pathway activity is inhibited by the tumor suppressor membrane protein, Patched. Hedgehog signaling is triggered by the secreted Hedgehog ligand, which binds and inhibits Patched, thus setting in motion the downstream events in signal transduction. Despite its critical importance, the mechanism by which Hedgehog antagonizes Patched has remained unknown. Here, we show that vertebrate Patched1 inhibition is caused by direct, palmitate-dependent interaction with the Sonic Hedgehog ligand. We find that a short palmitoylated N-terminal fragment of Sonic Hedgehog binds Patched1 and, strikingly, is sufficient to inhibit it and to activate signaling. The rest of Sonic Hedgehog confers high-affinity Patched1 binding and internalization through a distinct binding site, but, surprisingly, it is not absolutely required for signaling. The palmitate-dependent interaction with Patched1 is specifically impaired in a Sonic Hedgehog mutant causing human holoprosencephaly, the most frequent congenital brain malformation, explaining its drastically reduced potency. The palmitate-dependent interaction is also abolished in constitutively inhibited Patched1 point mutants causing the Gorlin cancer syndrome, suggesting that they might adopt a conformation distinct from the wild type. Our data demonstrate that Sonic Hedgehog signals via the palmitate-dependent arm of a two-pronged contact with Patched1. Furthermore, our results suggest that, during Hedgehog signaling, ligand binding inhibits Patched by trapping it in an inactive conformation, a mechanism that explains the dramatically reduced activity of oncogenic Patched1 mutants.
Collapse
|
23
|
Resh MD. Fatty acylation of proteins: The long and the short of it. Prog Lipid Res 2016; 63:120-31. [PMID: 27233110 DOI: 10.1016/j.plipres.2016.05.002] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/19/2016] [Accepted: 05/21/2016] [Indexed: 12/22/2022]
Abstract
Long, short and medium chain fatty acids are covalently attached to hundreds of proteins. Each fatty acid confers distinct biochemical properties, enabling fatty acylation to regulate intracellular trafficking, subcellular localization, protein-protein and protein-lipid interactions. Myristate and palmitate represent the most common fatty acid modifying groups. New insights into how fatty acylation reactions are catalyzed, and how fatty acylation regulates protein structure and function continue to emerge. Myristate is typically linked to an N-terminal glycine, but recent studies reveal that lysines can also be myristoylated. Enzymes that remove N-terminal myristoyl-glycine or myristate from lysines have now been identified. DHHC proteins catalyze S-palmitoylation, but the mechanisms that regulate substrate recognition by individual DHHC family members remain to be determined. New studies continue to reveal thioesterases that remove palmitate from S-acylated proteins. Another area of rapid expansion is fatty acylation of the secreted proteins hedgehog, Wnt and Ghrelin, by Hhat, Porcupine and GOAT, respectively. Understanding how these membrane bound O-acyl transferases recognize their protein and fatty acyl CoA substrates is an active area of investigation, and is punctuated by the finding that these enzymes are potential drug targets in human diseases.
Collapse
Affiliation(s)
- Marilyn D Resh
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 143, New York, NY 10075, United States.
| |
Collapse
|
24
|
Jakobs P, Schulz P, Ortmann C, Schürmann S, Exner S, Rebollido-Rios R, Dreier R, Seidler DG, Grobe K. Bridging the gap: heparan sulfate and Scube2 assemble Sonic hedgehog release complexes at the surface of producing cells. Sci Rep 2016; 6:26435. [PMID: 27199253 PMCID: PMC4873810 DOI: 10.1038/srep26435] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/03/2016] [Indexed: 12/12/2022] Open
Abstract
Decision making in cellular ensembles requires the dynamic release of signaling molecules from the producing cells into the extracellular compartment. One important example of molecules that require regulated release in order to signal over several cell diameters is the Hedgehog (Hh) family, because all Hhs are synthesized as dual-lipidated proteins that firmly tether to the outer membrane leaflet of the cell that produces them. Factors for the release of the vertebrate Hh family member Sonic Hedgehog (Shh) include cell-surface sheddases that remove the lipidated terminal peptides, as well as the soluble glycoprotein Scube2 that cell-nonautonomously enhances this process. This raises the question of how soluble Scube2 is recruited to cell-bound Shh substrates to regulate their turnover. We hypothesized that heparan sulfate (HS) proteoglycans (HSPGs) on the producing cell surface may play this role. In this work, we confirm that HSPGs enrich Scube2 at the surface of Shh-producing cells and that Scube2-regulated proteolytic Shh processing and release depends on specific HS. This finding indicates that HSPGs act as cell-surface assembly and storage platforms for Shh substrates and for protein factors required for their release, making HSPGs critical decision makers for Scube2-dependent Shh signaling from the surface of producing cells.
Collapse
Affiliation(s)
- P Jakobs
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - P Schulz
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - C Ortmann
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - S Schürmann
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - S Exner
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - R Rebollido-Rios
- Center for Medical Biotechnology#, University of Duisburg-Essen, 45117 Essen, Germany
| | - R Dreier
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| | - D G Seidler
- Centre for Internal Medicine, Hannover Medical School I3, EB2/R3110, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - K Grobe
- Institute for Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Waldeyerstr. 15, D-48149 Münster, Germany
| |
Collapse
|
25
|
Gauron C, Meda F, Dupont E, Albadri S, Quenech'Du N, Ipendey E, Volovitch M, Del Bene F, Joliot A, Rampon C, Vriz S. Hydrogen peroxide (H2O2) controls axon pathfinding during zebrafish development. Dev Biol 2016; 414:133-41. [PMID: 27158028 DOI: 10.1016/j.ydbio.2016.05.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 01/20/2023]
Abstract
It is now becoming evident that hydrogen peroxide (H2O2), which is constantly produced by nearly all cells, contributes to bona fide physiological processes. However, little is known regarding the distribution and functions of H2O2 during embryonic development. To address this question, we used a dedicated genetic sensor and revealed a highly dynamic spatio-temporal pattern of H2O2 levels during zebrafish morphogenesis. The highest H2O2 levels are observed during somitogenesis and organogenesis, and these levels gradually decrease in the mature tissues. Biochemical and pharmacological approaches revealed that H2O2 distribution is mainly controlled by its enzymatic degradation. Here we show that H2O2 is enriched in different regions of the developing brain and demonstrate that it participates to axonal guidance. Retinal ganglion cell axonal projections are impaired upon H2O2 depletion and this defect is rescued by H2O2 or ectopic activation of the Hedgehog pathway. We further show that ex vivo, H2O2 directly modifies Hedgehog secretion. We propose that physiological levels of H2O2 regulate RGCs axonal growth through the modulation of Hedgehog pathway.
Collapse
Affiliation(s)
- Carole Gauron
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; PSL Research University, 75005 Paris, France
| | - Francesca Meda
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; École Normale Supérieure, Institute of Biology at the Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, F-75005 Paris, France; PSL Research University, 75005 Paris, France
| | - Edmond Dupont
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; PSL Research University, 75005 Paris, France
| | - Shahad Albadri
- Institut Curie CNRS UMR3215, INSERM U934, F-75248, France; PSL Research University, 75005 Paris, France
| | - Nicole Quenech'Du
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; PSL Research University, 75005 Paris, France
| | - Eliane Ipendey
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; École Normale Supérieure, Institute of Biology at the Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, F-75005 Paris, France; PSL Research University, 75005 Paris, France
| | - Michel Volovitch
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; École Normale Supérieure, Institute of Biology at the Ecole Normale Supérieure (IBENS), CNRS UMR8197, INSERM U1024, F-75005 Paris, France; PSL Research University, 75005 Paris, France
| | - Filippo Del Bene
- Institut Curie CNRS UMR3215, INSERM U934, F-75248, France; PSL Research University, 75005 Paris, France
| | - Alain Joliot
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; PSL Research University, 75005 Paris, France
| | - Christine Rampon
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; Université Paris Diderot, Sorbonne Paris Cité, Biology Department, 75205 Paris Cedex 13, France; PSL Research University, 75005 Paris, France
| | - Sophie Vriz
- Centre Interdisciplinaire de Recherche en Biologie (CIRB), CNRS UMR 7241/INSERM U1050/Collège de France, 11, Place Marcelin Berthelot, 75231 Paris Cedex 05, France; Université Paris Diderot, Sorbonne Paris Cité, Biology Department, 75205 Paris Cedex 13, France; PSL Research University, 75005 Paris, France.
| |
Collapse
|
26
|
Nile AH, Hannoush RN. Fatty acylation of Wnt proteins. Nat Chem Biol 2016; 12:60-9. [DOI: 10.1038/nchembio.2005] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/09/2015] [Indexed: 02/04/2023]
|
27
|
Fatty acyl donor selectivity in membrane bound O-acyltransferases and communal cell fate decision-making. Biochem Soc Trans 2016; 43:235-9. [PMID: 25849923 DOI: 10.1042/bst20140282] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The post-translational modification of proteins with lipid moieties confers spatial and temporal control of protein function by restricting their subcellular distribution or movement in the extracellular milieu. Yet, little is known about the significance of lipid selectivity to the activity of proteins targeted for such modifications. Membrane bound O-acyl transferases (MBOATs) are a superfamily of multipass enzymes that transfer fatty acids on to lipid or protein substrates. Three MBOATs constitute a subfamily with secreted signalling molecules for substrates, the Wnt, Hedgehog (Hh) and Ghrelin proteins. Given their important roles in adult tissue homoeostasis, all three molecules and their respective associated acyltransferases provide a framework for interrogating the role of extracellular acylation events in cell-to-cell communication. Here, we discuss how the preference for a fatty acyl donor in the Wnt acyltransferase porcupine (Porcn) and possibly in other protein lipidation enzymes may provide a means for coupling metabolic health at the single cell level to communal cell fate decision-making in complex multicellular organisms.
Collapse
|
28
|
Lanyon-Hogg T, Masumoto N, Bodakh G, Konitsiotis AD, Thinon E, Rodgers UR, Owens RJ, Magee AI, Tate EW. Click chemistry armed enzyme-linked immunosorbent assay to measure palmitoylation by hedgehog acyltransferase. Anal Biochem 2015; 490:66-72. [PMID: 26334609 PMCID: PMC4615133 DOI: 10.1016/j.ab.2015.08.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 12/30/2022]
Abstract
Hedgehog signaling is critical for correct embryogenesis and tissue development. However, on maturation, signaling is also found to be aberrantly activated in many cancers. Palmitoylation of the secreted signaling protein sonic hedgehog (Shh) by the enzyme hedgehog acyltransferase (Hhat) is required for functional signaling. To quantify this important posttranslational modification, many in vitro Shh palmitoylation assays employ radiolabeled fatty acids, which have limitations in terms of cost and safety. Here we present a click chemistry armed enzyme-linked immunosorbent assay (click-ELISA) for assessment of Hhat activity through acylation of biotinylated Shh peptide with an alkyne-tagged palmitoyl-CoA (coenzyme A) analogue. Click chemistry functionalization of the alkyne tag with azido-FLAG peptide allows analysis through an ELISA protocol and colorimetric readout. This assay format identified the detergent n-dodecyl β-d-maltopyranoside as an improved solubilizing agent for Hhat activity. Quantification of the potency of RU-SKI small molecule Hhat inhibitors by click-ELISA indicated IC50 values in the low- or sub-micromolar range. A stopped assay format was also employed that allows measurement of Hhat kinetic parameters where saturating substrate concentrations exceed the binding capacity of the streptavidin-coated plate. Therefore, click-ELISA represents a nonradioactive method for assessing protein palmitoylation in vitro that is readily expandable to other classes of protein lipidation.
Collapse
Affiliation(s)
- Thomas Lanyon-Hogg
- Institute of Chemical Biology, Department of Chemistry, Imperial College London, South Kensington SW7 2AZ, UK
| | - Naoko Masumoto
- Institute of Chemical Biology, Department of Chemistry, Imperial College London, South Kensington SW7 2AZ, UK
| | - George Bodakh
- Institute of Chemical Biology, Department of Chemistry, Imperial College London, South Kensington SW7 2AZ, UK
| | - Antonio D Konitsiotis
- Molecular Medicine Section, National Heart & Lung Institute, Imperial College London, South Kensington SW7 2AZ, UK
| | - Emmanuelle Thinon
- Institute of Chemical Biology, Department of Chemistry, Imperial College London, South Kensington SW7 2AZ, UK
| | - Ursula R Rodgers
- Molecular Medicine Section, National Heart & Lung Institute, Imperial College London, South Kensington SW7 2AZ, UK
| | - Raymond J Owens
- Division of Structural Biology, Henry Wellcome Building for Genomic Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK; OPPF-UK, The Research Complex at Harwell, Rutherford Appleton Laboratory, Harwell, Oxfordshire, OX11 0FA, UK
| | - Anthony I Magee
- Molecular Medicine Section, National Heart & Lung Institute, Imperial College London, South Kensington SW7 2AZ, UK.
| | - Edward W Tate
- Institute of Chemical Biology, Department of Chemistry, Imperial College London, South Kensington SW7 2AZ, UK.
| |
Collapse
|
29
|
Targeting the Hedgehog Pathway in Pediatric Medulloblastoma. Cancers (Basel) 2015; 7:2110-23. [PMID: 26512695 PMCID: PMC4695880 DOI: 10.3390/cancers7040880] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/05/2015] [Accepted: 10/16/2015] [Indexed: 11/17/2022] Open
Abstract
Medulloblastoma (MB), a primitive neuroectomal tumor of the cerebellum, is the most common malignant pediatric brain tumor. The cause of MB is largely unknown, but aberrant activation of Hedgehog (Hh) pathway is responsible for ~30% of MB. Despite aggressive treatment with surgical resection, radiation and chemotherapy, 70%–80% of pediatric medulloblastoma cases can be controlled, but most treated patients suffer devastating side effects. Therefore, developing a new effective treatment strategy is urgently needed. Hh signaling controls transcription of target genes by regulating activities of the three Glioma-associated oncogene (Gli1-3) transcription factors. In this review, we will focus on current clinical treatment options of MB and discuss mechanisms of drug resistance. In addition, we will describe current known molecular pathways which crosstalk with the Hedgehog pathway both in the context of medulloblastoma and non-medulloblastoma cancer development. Finally, we will introduce post-translational modifications that modulate Gli1 activity and summarize the positive and negative regulations of the Hh/Gli1 pathway. Towards developing novel combination therapies for medulloblastoma treatment, current information on interacting pathways and direct regulation of Hh signaling should prove critical.
Collapse
|
30
|
Ortmann C, Pickhinke U, Exner S, Ohlig S, Lawrence R, Jboor H, Dreier R, Grobe K. Sonic hedgehog processing and release are regulated by glypican heparan sulfate proteoglycans. J Cell Sci 2015; 128:2374-85. [PMID: 25967551 DOI: 10.1242/jcs.170670] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/05/2015] [Indexed: 12/21/2022] Open
Abstract
All Hedgehog morphogens are released from producing cells, despite being synthesized as N- and C-terminally lipidated molecules, a modification that firmly tethers them to the cell membrane. We have previously shown that proteolytic removal of both lipidated peptides, called shedding, releases bioactive Sonic hedgehog (Shh) morphogens from the surface of transfected Bosc23 cells. Using in vivo knockdown together with in vitro cell culture studies, we now show that glypican heparan sulfate proteoglycans regulate this process, through their heparan sulfate chains, in a cell autonomous manner. Heparan sulfate specifically modifies Shh processing at the cell surface, and purified glycosaminoglycans enhance the proteolytic removal of N- and C-terminal Shh peptides under cell-free conditions. The most likely explanation for these observations is direct Shh processing in the extracellular compartment, suggesting that heparan sulfate acts as a scaffold or activator for Shh ligands and the factors required for their turnover. We also show that purified heparan sulfate isolated from specific cell types and tissues mediates the release of bioactive Shh from pancreatic cancer cells, revealing a previously unknown regulatory role for these versatile molecules in a pathological context.
Collapse
Affiliation(s)
- Corinna Ortmann
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Ute Pickhinke
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Sebastian Exner
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Stefanie Ohlig
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Roger Lawrence
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Hamodah Jboor
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Rita Dreier
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| | - Kay Grobe
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, 48149 Münster, Germany
| |
Collapse
|
31
|
Darling JE, Zhao F, Loftus RJ, Patton LM, Gibbs RA, Hougland JL. Structure–Activity Analysis of Human Ghrelin O-Acyltransferase Reveals Chemical Determinants of Ghrelin Selectivity and Acyl Group Recognition. Biochemistry 2015; 54:1100-10. [DOI: 10.1021/bi5010359] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Joseph E. Darling
- Department
of Chemistry, Syracuse University, Syracuse, New York 13244, United States
| | - Feifei Zhao
- Department
of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Rosemary J. Loftus
- Department
of Chemistry, Syracuse University, Syracuse, New York 13244, United States
| | - Leslie M. Patton
- Department
of Chemistry, Syracuse University, Syracuse, New York 13244, United States
| | - Richard A. Gibbs
- Department
of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - James L. Hougland
- Department
of Chemistry, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
32
|
Petrova E, Matevossian A, Resh MD. Hedgehog acyltransferase as a target in pancreatic ductal adenocarcinoma. Oncogene 2015; 34:263-8. [PMID: 24469057 PMCID: PMC4513646 DOI: 10.1038/onc.2013.575] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 11/20/2013] [Accepted: 11/29/2013] [Indexed: 12/24/2022]
Abstract
Sonic Hedgehog (Shh) is abnormally expressed in pancreatic cancer and is associated with disease onset and progression. Inhibition of Shh signaling is thus an attractive clinical target for therapeutic intervention. Most efforts to block Shh signaling have focused on inhibitors of Smoothened, which target the canonical Shh signaling pathway. These approaches have met with limited success, in part due to development of resistance-conferring mutations and contributions from non-canonical signaling pathways. Here, we show that Hedgehog acyltransferase (Hhat), the enzyme responsible for the attachment of palmitate onto Shh, is a novel target for inhibition of Shh signaling in pancreatic cancer cells. Depletion of Hhat with lentivirally delivered small hairpin RNA decreased both anchorage-dependent and independent proliferation of human pancreatic cancer cells. In vivo, Hhat knockdown led to reduction of tumor growth in a mouse xenograft model of pancreatic cancer. RU-SKI 43, a small molecule inhibitor of Hhat recently developed by our group, reduced pancreatic cancer cell proliferation and Gli-1 activation through Smoothened-independent non-canonical signaling. In addition, RU-SKI 43 treatment inhibited two key proliferative pathways regulated by Akt and mTOR. This work demonstrates that Hhat has a critical role in pancreatic cancer and that a small molecule inhibitor of Hhat can successfully block pancreatic cancer cell proliferation. It also highlights the importance of developing optimized Hhat inhibitors to be used as therapeutics in pancreatic cancer, as well as in other malignancies characterized by Shh overexpression.
Collapse
Affiliation(s)
- E Petrova
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Graduate Program in Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - A Matevossian
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Gerstner Sloan-Kettering Graduate School of Biomedical Sciences, New York, NY, USA
| | - MD Resh
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Gerstner Sloan-Kettering Graduate School of Biomedical Sciences, New York, NY, USA
- Graduate Programs in Cell Biology and Biochemistry, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| |
Collapse
|
33
|
Miranda M, Galli LM, Enriquez M, Szabo LA, Gao X, Hannoush RN, Burrus LW. Identification of the WNT1 residues required for palmitoylation by Porcupine. FEBS Lett 2014; 588:4815-24. [PMID: 25451226 DOI: 10.1016/j.febslet.2014.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 10/27/2014] [Accepted: 11/12/2014] [Indexed: 11/26/2022]
Abstract
The post-translational palmitoylation of WNT morphogens is critical for proper signaling during embryogenesis and adult homeostasis. The addition of palmitoyl groups to WNT proteins is catalyzed by Porcupine (PORCN). However, the Wnt amino acid residues required for recognition and palmitoylation by PORCN have not been fully characterized. We show that WNT1 residues 214-234 are sufficient for PORCN-dependent palmitoylation of Ser224. Substitution of Ser224 with Thr, but not Cys, is tolerated in palmitoylation and biological assays. Our data highlight the importance of palmitoylation for WNT1 activity and establish PORCN as an O-acyl transferase for WNT1.
Collapse
Affiliation(s)
- M Miranda
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - L M Galli
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - M Enriquez
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - L A Szabo
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - X Gao
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - R N Hannoush
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - L W Burrus
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA.
| |
Collapse
|
34
|
Adams ST, Miller SC. Beyond D-luciferin: expanding the scope of bioluminescence imaging in vivo. Curr Opin Chem Biol 2014; 21:112-20. [PMID: 25078002 DOI: 10.1016/j.cbpa.2014.07.003] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/28/2014] [Accepted: 07/03/2014] [Indexed: 01/10/2023]
Abstract
The light-emitting chemical reaction catalyzed by the enzyme firefly luciferase is widely used for noninvasive imaging in live mice. However, photon emission from the luciferase is crucially dependent on the chemical properties of its substrate, D-luciferin. In this review, we describe recent work to replace the natural luciferase substrate with synthetic analogs that extend the scope of bioluminescence imaging.
Collapse
Affiliation(s)
- Spencer T Adams
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Stephen C Miller
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
35
|
Rios-Esteves J, Haugen B, Resh MD. Identification of key residues and regions important for porcupine-mediated Wnt acylation. J Biol Chem 2014; 289:17009-19. [PMID: 24798332 DOI: 10.1074/jbc.m114.561209] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Wnts comprise a family of lipid-modified, secreted signaling proteins that control embryogenesis, as well as tissue homeostasis in adults. Post-translational attachment of palmitoleate (C16:1) to a conserved Ser in Wnt proteins is catalyzed by Porcupine (Porcn), a member of the membrane bound O-acyltransferase (MBOAT) family, and is required for Wnt secretion and signaling. Moreover, genetic alterations in the PORCN gene lead to focal dermal hypoplasia, an X-linked developmental disorder. Despite its physiological importance, the biochemical mechanism governing Wnt acylation by Porcn is poorly understood. Here, we use a cell-based fatty acylation assay that is a direct readout of Porcn acyltransferase activity to perform structure-function analysis of highly conserved residues in Porcn and Wnt3a. In total, 16-point mutations in Porcn and 13 mutations in Wnt3a were generated and analyzed. We identified key residues within Porcn required for enzymatic activity, stability, and Wnt3a binding and mapped these active site residues to predicted transmembrane domain 9. Analysis of focal dermal hypoplasia-associated mutations in Porcn revealed that loss of enzymatic activity arises from altered stability. A consensus sequence within Wnt3a was identified (CXCHGXSXXCXXKXC) that contains residues that mediate Porcn binding, fatty acid transfer, and Wnt signaling. We also showed that Ser or Thr, but not Cys, can serve as a fatty acylation site in Wnt, establishing Porcn as an O-acyltransferase. This analysis sheds light into the mechanism by which Porcn transfers fatty acids to Wnt proteins and provides insight into the mechanisms of fatty acid transfer by MBOAT family members.
Collapse
Affiliation(s)
- Jessica Rios-Esteves
- From the Cell Biology Program and the Gerstner Sloan-Kettering Graduate School of Biomedical Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | - Brittany Haugen
- From the Cell Biology Program and the Gerstner Sloan-Kettering Graduate School of Biomedical Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | - Marilyn D Resh
- From the Cell Biology Program and the Gerstner Sloan-Kettering Graduate School of Biomedical Sciences, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
36
|
Jakobs P, Exner S, Schürmann S, Pickhinke U, Bandari S, Ortmann C, Kupich S, Schulz P, Hansen U, Seidler DG, Grobe K. Scube2 enhances proteolytic Shh processing from the surface of Shh-producing cells. J Cell Sci 2014; 127:1726-37. [PMID: 24522195 DOI: 10.1242/jcs.137695] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
All morphogens of the Hedgehog (Hh) family are synthesized as dual-lipidated proteins, which results in their firm attachment to the surface of the cell in which they were produced. Thus, Hh release into the extracellular space requires accessory protein activities. We suggested previously that the proteolytic removal of N- and C-terminal lipidated peptides (shedding) could be one such activity. More recently, the secreted glycoprotein Scube2 (signal peptide, cubulin domain, epidermal-growth-factor-like protein 2) was also implicated in the release of Shh from the cell membrane. This activity strictly depended on the CUB domains of Scube2, which derive their name from the complement serine proteases and from bone morphogenetic protein-1/tolloid metalloproteinases (C1r/C1s, Uegf and Bmp1). CUB domains function as regulators of proteolytic activity in these proteins. This suggested that sheddases and Scube2 might cooperate in Shh release. Here, we confirm that sheddases and Scube2 act cooperatively to increase the pool of soluble bioactive Shh, and that Scube2-dependent morphogen release is unequivocally linked to the proteolytic processing of lipidated Shh termini, resulting in truncated soluble Shh. Thus, Scube2 proteins act as protease enhancers in this setting, revealing newly identified Scube2 functions in Hh signaling regulation.
Collapse
Affiliation(s)
- Petra Jakobs
- The Institute for Physiological Chemistry and Pathobiochemistry, Westfälische Wilhelms Universität Münster, Waldeyerstrasse 15, D-48149 Münster, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Taylor MS, Ruch TR, Hsiao PY, Hwang Y, Zhang P, Dai L, Huang CRL, Berndsen CE, Kim MS, Pandey A, Wolberger C, Marmorstein R, Machamer C, Boeke JD, Cole PA. Architectural organization of the metabolic regulatory enzyme ghrelin O-acyltransferase. J Biol Chem 2013; 288:32211-32228. [PMID: 24045953 DOI: 10.1074/jbc.m113.510313] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ghrelin O-acyltransferase (GOAT) is a polytopic integral membrane protein required for activation of ghrelin, a secreted metabolism-regulating peptide hormone. Although GOAT is a potential therapeutic target for the treatment of obesity and diabetes and plays a key role in other physiologic processes, little is known about its structure or mechanism. GOAT is a member of the membrane-bound O-acyltransferase (MBOAT) family, a group of polytopic integral membrane proteins involved in lipid-biosynthetic and lipid-signaling reactions from prokaryotes to humans. Here we use phylogeny and a variety of bioinformatic tools to predict the topology of GOAT. Using selective permeabilization indirect immunofluorescence microscopy in combination with glycosylation shift immunoblotting, we demonstrate that GOAT contains 11 transmembrane helices and one reentrant loop. Development of the V5Glyc tag, a novel, small, and sensitive dual topology reporter, facilitated these experiments. The MBOAT family invariant residue His-338 is in the ER lumen, consistent with other family members, but conserved Asn-307 is cytosolic, making it unlikely that both are involved in catalysis. Photocross-linking of synthetic ghrelin analogs and inhibitors demonstrates binding to the C-terminal region of GOAT, consistent with a role of His-338 in the active site. This knowledge of GOAT architecture is important for a deeper understanding of the mechanism of GOAT and other MBOATs and could ultimately advance the discovery of selective inhibitors for these enzymes.
Collapse
Affiliation(s)
- Martin S Taylor
- From the Department of Pharmacology and Molecular Sciences,; the High Throughput Biology Center and Department of Molecular Biology and Genetics
| | | | - Po-Yuan Hsiao
- From the Department of Pharmacology and Molecular Sciences
| | - Yousang Hwang
- From the Department of Pharmacology and Molecular Sciences
| | - Pingfeng Zhang
- the Program in Gene Expression and Regulation, The Wistar Institute, Philadelphia, Pennsylvania 19104
| | - Lixin Dai
- the High Throughput Biology Center and Department of Molecular Biology and Genetics
| | - Cheng Ran Lisa Huang
- the High Throughput Biology Center and Department of Molecular Biology and Genetics,; the McKusick-Nathans Institute of Genetic Medicine
| | - Christopher E Berndsen
- the Howard Hughes Medical Institute and Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Min-Sik Kim
- the McKusick-Nathans Institute of Genetic Medicine
| | | | - Cynthia Wolberger
- the Howard Hughes Medical Institute and Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Ronen Marmorstein
- the Program in Gene Expression and Regulation, The Wistar Institute, Philadelphia, Pennsylvania 19104; the Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | | - Jef D Boeke
- the High Throughput Biology Center and Department of Molecular Biology and Genetics,.
| | - Philip A Cole
- From the Department of Pharmacology and Molecular Sciences,.
| |
Collapse
|
38
|
Pan A, Chang L, Nguyen A, James AW. A review of hedgehog signaling in cranial bone development. Front Physiol 2013; 4:61. [PMID: 23565096 PMCID: PMC3613593 DOI: 10.3389/fphys.2013.00061] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/13/2013] [Indexed: 12/20/2022] Open
Abstract
During craniofacial development, the Hedgehog (HH) signaling pathway is essential for mesodermal tissue patterning and differentiation. The HH family consists of three protein ligands: Sonic Hedgehog (SHH), Indian Hedgehog (IHH), and Desert Hedgehog (DHH), of which two are expressed in the craniofacial complex (IHH and SHH). Dysregulations in HH signaling are well documented to result in a wide range of craniofacial abnormalities, including holoprosencephaly (HPE), hypotelorism, and cleft lip/palate. Furthermore, mutations in HH effectors, co-receptors, and ciliary proteins result in skeletal and craniofacial deformities. Cranial suture morphogenesis is a delicate developmental process that requires control of cell commitment, proliferation and differentiation. This review focuses on both what is known and what remains unknown regarding HH signaling in cranial suture morphogenesis and intramembranous ossification. As demonstrated from murine studies, expression of both SHH and IHH is critical to the formation and fusion of the cranial sutures and calvarial ossification. SHH expression has been observed in the cranial suture mesenchyme and its precise function is not fully defined, although some postulate SHH to delay cranial suture fusion. IHH expression is mainly found on the osteogenic fronts of the calvarial bones, and functions to induce cell proliferation and differentiation. Unfortunately, neonatal lethality of IHH deficient mice precludes a detailed examination of their postnatal calvarial phenotype. In summary, a number of basic questions are yet to be answered regarding domains of expression, developmental role, and functional overlap of HH morphogens in the calvaria. Nevertheless, SHH and IHH ligands are integral to cranial suture development and regulation of calvarial ossification. When HH signaling goes awry, the resultant suite of morphologic abnormalities highlights the important roles of HH signaling in cranial development.
Collapse
Affiliation(s)
- Angel Pan
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | |
Collapse
|