1
|
Zhong Y, Chen X, Wu S, Fang H, Hong L, Shao L, Wang L, Wu J. Deciphering colorectal cancer radioresistance and immune microrenvironment: unraveling the role of EIF5A through single-cell RNA sequencing and machine learning. Front Immunol 2024; 15:1466226. [PMID: 39290702 PMCID: PMC11405197 DOI: 10.3389/fimmu.2024.1466226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Background Radiotherapy (RT) is a critical component of treatment for locally advanced rectal cancer (LARC), though patient response varies significantly. The variability in treatment outcomes is partly due to the resistance conferred by cancer stem cells (CSCs) and tumor immune microenvironment (TiME). This study investigates the role of EIF5A in radiotherapy response and its impact on the CSCs and TiME. Methods Predictive models for preoperative radiotherapy (preRT) response were developed using machine learning, identifying EIF5A as a key gene associated with radioresistance. EIF5A expression was analyzed via bulk RNA-seq and single-cell RNA-seq (scRNA-seq). Functional assays and in vivo experiments validated EIF5A's role in radioresistance and TiME modulation. Results EIF5A was significantly upregulated in radioresistant colorectal cancer (CRC) tissues. EIF5A knockdown in CRC cell lines reduced cell viability, migration, and invasion after radiation, and increased radiation-induced apoptosis. Mechanistically, EIF5A promoted cancer stem cell (CSC) characteristics through the Hedgehog signaling pathway. Analysis of the TiME revealed that the radiation-resistant group had an immune-desert phenotype, characterized by low immune cell infiltration. In vivo experiments showed that EIF5A knockdown led to increased infiltration of CD8+ T cells and M1 macrophages, and decreased M2 macrophages and Tregs following radiation therapy, thereby enhancing the radiotherapy response. Conclusion EIF5A contributes to CRC radioresistance by promoting CSC traits via the Hedgehog pathway and modulating the TiME to an immune-suppressive state. Targeting EIF5A could enhance radiation sensitivity and improve immune responses, offering a potential therapeutic strategy to optimize radiotherapy outcomes in CRC patients.
Collapse
Affiliation(s)
- Yaqi Zhong
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University Fujian Cancer Hospital, (Fujian Branch of Fudan University Shanghai Cancer Center), Fujian Cancer Hospital, Fuzhou, China
| | - Xingte Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University Fujian Cancer Hospital, (Fujian Branch of Fudan University Shanghai Cancer Center), Fujian Cancer Hospital, Fuzhou, China
| | - Shiji Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University Fujian Cancer Hospital, (Fujian Branch of Fudan University Shanghai Cancer Center), Fujian Cancer Hospital, Fuzhou, China
| | - Huipeng Fang
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Liang Hong
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University Fujian Cancer Hospital, (Fujian Branch of Fudan University Shanghai Cancer Center), Fujian Cancer Hospital, Fuzhou, China
| | - Lingdong Shao
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University Fujian Cancer Hospital, (Fujian Branch of Fudan University Shanghai Cancer Center), Fujian Cancer Hospital, Fuzhou, China
| | - Lei Wang
- Department of Radiation Oncology, Jiangxi Clinical Research Center for Cancer, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Junxin Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University Fujian Cancer Hospital, (Fujian Branch of Fudan University Shanghai Cancer Center), Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
2
|
Kulkarni PP, Ekhlak M, Dash D. Non-canonical non-genomic morphogen signaling in anucleate platelets: a critical determinant of prothrombotic function in circulation. Cell Commun Signal 2024; 22:13. [PMID: 38172855 PMCID: PMC10763172 DOI: 10.1186/s12964-023-01448-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Circulating platelets derived from bone marrow megakaryocytes play a central role in thrombosis and hemostasis. Despite being anucleate, platelets express several proteins known to have nuclear niche. These include transcription factors and steroid receptors whose non-genomic functions are being elucidated in platelets. Quite remarkably, components of some of the best-studied morphogen pathways, namely Notch, Sonic Hedgehog (Shh), and Wnt have also been described in recent years in platelets, which regulate platelet function in the context of thrombosis as well as influence their survival. Shh and Notch pathways in stimulated platelets establish feed-forward loops of autocrine/juxtacrine/paracrine non-canonical signaling that helps perpetuate thrombosis. On the other hand, non-canonical Wnt signaling is part of a negative feedback loop for restricting platelet activation and possibly limiting thrombus growth. The present review will provide an overview of these signaling pathways in general. We will then briefly discuss the non-genomic roles of transcription factors and steroid receptors in platelet activation. This will be followed by an elaborate description of morphogen signaling in platelets with a focus on their bearing on platelet activation leading to hemostasis and thrombosis as well as their potential for therapeutic targeting in thrombotic disorders.
Collapse
Affiliation(s)
- Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
3
|
Feng Z, Zhu S, Li W, Yao M, Song H, Wang RB. Current approaches and strategies to identify Hedgehog signaling pathway inhibitors for cancer therapy. Eur J Med Chem 2022; 244:114867. [DOI: 10.1016/j.ejmech.2022.114867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022]
|
4
|
Zhou M, Han Y, Wang B, Cho YS, Jiang J. Dose-dependent phosphorylation and activation of Hh pathway transcription factors. Life Sci Alliance 2022; 5:5/11/e202201570. [PMID: 36271509 PMCID: PMC9445324 DOI: 10.26508/lsa.202201570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Graded Hedgehog (Hh) signaling is mediated by graded Cubitus interruptus (Ci)/Gli transcriptional activity, but how the Hh gradient is converted into the Ci/Gli activity gradient remains poorly understood. Here, we show that graded Hh induces a progressive increase in Ci phosphorylation at multiple Fused (Fu)/CK1 sites including a cluster located in the C-terminal Sufu-binding domain. We demonstrated that Fu directly phosphorylated Ci on S1382, priming CK1 phosphorylation on adjacent sites, and that Fu/CK1-mediated phosphorylation of the C-terminal sites interfered with Sufu binding and facilitated Ci activation. Phosphorylation at the N-terminal, middle, and C-terminal Fu/CK1 sites occurred independently of one another and each increased progressively in response to increasing levels of Hh or increasing amounts of Hh exposure time. Increasing the number of phospho-mimetic mutations of Fu/CK1 sites resulted in progressively increased Ci activation by alleviating Sufu-mediated inhibition. We found that the C-terminal Fu/CK1 phosphorylation cluster is conserved in Gli2 and contributes to its dose-dependent activation. Our study suggests that the Hh signaling gradient is translated into a Ci/Gli phosphorylation gradient that activates Ci/Gli by gradually releasing Sufu-mediated inhibition.
Collapse
Affiliation(s)
- Mengmeng Zhou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuhong Han
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bing Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yong Suk Cho
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA .,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
Jiang J. Hedgehog signaling mechanism and role in cancer. Semin Cancer Biol 2022; 85:107-122. [PMID: 33836254 PMCID: PMC8492792 DOI: 10.1016/j.semcancer.2021.04.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/25/2021] [Accepted: 04/02/2021] [Indexed: 12/12/2022]
Abstract
Cell-cell communication through evolutionarily conserved signaling pathways governs embryonic development and adult tissue homeostasis. Deregulation of these signaling pathways has been implicated in a wide range of human diseases including cancer. One such pathway is the Hedgehog (Hh) pathway, which was originally discovered in Drosophila and later found to play a fundamental role in human development and diseases. Abnormal Hh pathway activation is a major driver of basal cell carcinomas (BCC) and medulloblastoma. Hh exerts it biological influence through a largely conserved signal transduction pathway from the activation of the GPCR family transmembrane protein Smoothened (Smo) to the conversion of latent Zn-finger transcription factors Gli/Ci proteins from their repressor (GliR/CiR) to activator (GliA/CiA) forms. Studies from model organisms and human patients have provided deep insight into the Hh signal transduction mechanisms, revealed roles of Hh signaling in a wide range of human cancers, and suggested multiple strategies for targeting this pathway in cancer treatment.
Collapse
Affiliation(s)
- Jin Jiang
- Department of Molecular Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
| |
Collapse
|
6
|
Gonçalves Antunes M, Sanial M, Contremoulins V, Carvalho S, Plessis A, Becam I. High hedgehog signaling is transduced by a multikinase-dependent switch controlling the apico-basal distribution of the GPCR smoothened. eLife 2022; 11:79843. [PMID: 36083801 PMCID: PMC9462849 DOI: 10.7554/elife.79843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The oncogenic G-protein-coupled receptor (GPCR) Smoothened (SMO) is a key transducer of the hedgehog (HH) morphogen, which plays an essential role in the patterning of epithelial structures. Here, we examine how HH controls SMO subcellular localization and activity in a polarized epithelium using the Drosophila wing imaginal disc as a model. We provide evidence that HH promotes the stabilization of SMO by switching its fate after endocytosis toward recycling. This effect involves the sequential and additive action of protein kinase A, casein kinase I, and the Fused (FU) kinase. Moreover, in the presence of very high levels of HH, the second effect of FU leads to the local enrichment of SMO in the most basal domain of the cell membrane. Together, these results link the morphogenetic effects of HH to the apico-basal distribution of SMO and provide a novel mechanism for the regulation of a GPCR.
Collapse
Affiliation(s)
| | | | | | | | - Anne Plessis
- Université Paris Cité, CNRS, Institut Jacques Monod
| | | |
Collapse
|
7
|
Wang W, Shiraishi R, Kawauchi D. Sonic Hedgehog Signaling in Cerebellar Development and Cancer. Front Cell Dev Biol 2022; 10:864035. [PMID: 35573667 PMCID: PMC9100414 DOI: 10.3389/fcell.2022.864035] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/28/2022] [Indexed: 12/30/2022] Open
Abstract
The sonic hedgehog (SHH) pathway regulates the development of the central nervous system in vertebrates. Aberrant regulation of SHH signaling pathways often causes neurodevelopmental diseases and brain tumors. In the cerebellum, SHH secreted by Purkinje cells is a potent mitogen for granule cell progenitors, which are the most abundant cell type in the mature brain. While a reduction in SHH signaling induces cerebellar structural abnormalities, such as hypoplasia in various genetic disorders, the constitutive activation of SHH signaling often induces medulloblastoma (MB), one of the most common pediatric malignant brain tumors. Based on the existing literature on canonical and non-canonical SHH signaling pathways, emerging basic and clinical studies are exploring novel therapeutic approaches for MB by targeting SHH signaling at distinct molecular levels. In this review, we discuss the present consensus on SHH signaling mechanisms, their roles in cerebellar development and tumorigenesis, and the recent advances in clinical trials for MB.
Collapse
Affiliation(s)
- Wanchen Wang
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Ryo Shiraishi
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
- Department of NCNP Brain Physiology and Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Kawauchi
- Department of Biochemistry and Cellular Biology, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
- *Correspondence: Daisuke Kawauchi,
| |
Collapse
|
8
|
Jia J, Jiang J. Regulation of Smoothened Trafficking and Abundance in Hedgehog Signaling. Front Cell Dev Biol 2022; 10:847844. [PMID: 35321245 PMCID: PMC8936432 DOI: 10.3389/fcell.2022.847844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/23/2022] [Indexed: 12/02/2022] Open
Abstract
The GPCR-family protein Smoothened (Smo) is essential for Hedgehog (Hh) signal transduction in both insects and vertebrates. The regulation of subcellular localization and abundance of Smo is a critical step in Hh signaling. Recent studies have demonstrated that Smo is subjected to ubiquitination mediated by multiple E3 ubiquitin ligases, leading to Smo endocytosis and subsequent degradation through the proteasome- and lysosome-mediated pathways in Drosophila. Ubiquitination of Smo also promotes its ciliary exit in mammalian cells. Hh inhibits Smo ubiquitination by blocking E3 ligase recruitment and promoting Smo deubiquitination through the ubiquitin-specific protease 8 (USP8) in Drosophila. Inhibition of Smo ubiquitination by Hh promotes Smo cell surface accumulation in Drosophila and ciliary accumulation in mammalian cells. Interestingly, Hh also induces sumoylation of Smo in both Drosophila and mammalian cells, which promotes Smo cell surface/ciliary accumulation. This review focuses on how ubiquitination and sumoylation regulate Smo intracellular trafficking and abundance and how these processes are regulated by Hh.
Collapse
Affiliation(s)
- Jianhang Jia
- Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jin Jiang
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
9
|
Characterization of Smoothened Phosphorylation and Activation. Methods Mol Biol 2022; 2374:121-137. [PMID: 34562248 PMCID: PMC8941978 DOI: 10.1007/978-1-0716-1701-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The GPCR-family protein Smoothened (Smo) is an obligatory signal transducer of the Hedgehog (Hh) signaling pathway. Binding of Hh to its receptor Patched (Ptc) alleviates Ptc-mediated inhibition of Smo, allowing Smo to activate the Cubitus interruptus (Ci)/Gli family of zinc finger transcription factors. The activation of Smo is an early and crucial event in Hh signal transduction. Studies have shown that Hh induces cell surface/ciliary accumulation and phosphorylation of Smo by multiple kinases, including protein kinase A (PKA), casein kinase 1 (CK1), casein kinase 2 (CK2), G protein-coupled receptor kinase 2 (Gprk2/GRK2), and atypical PKC (aPKC). Here, we describe the assays used to examine the phosphorylation and activity of Smo, including in vitro kinase assay, phospho-specific antibodies, luciferase reporter assay, cell surface accumulation, and ciliary localization assays. These assays provide powerful tools to study Smo phosphorylation and activation, leading to mechanistic insight into Smo regulation.
Collapse
|
10
|
Abstract
The Hedgehog (Hh) family of secreted proteins governs embryonic development and adult tissue homeostasis by regulating the abundance, localization, and activity of the GPCR family protein Smoothened (Smo). Smo trafficking and subcellular accumulation are controlled by multiple posttranslational modifications (PTMs) including phosphorylation, ubiquitination, and sumoylation, which appears to be conserved from Drosophila to mammals. Smo ubiquitination is dynamically regulated by E3 ubiquitin ligases and deubiquitinases (dubs) and is opposed by Hh signaling. By contrast, Smo sumoylation is stimulated by Hh, which counteracts Smo ubiquitination by recruiting the dub USP8. We describe cell-base assays for Smo ubiquitination and its regulation by Hh and the E3 ligases in Drosophila. We also describe assays for Smo sumoylation in both Drosophila and mammalian cultured cells.
Collapse
Affiliation(s)
- Yuhong Han
- Department of Molecular Biology, UT Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Jin Jiang
- Department of Molecular Biology, UT Southwestern Medical Center at Dallas, Dallas, TX, USA.
| |
Collapse
|
11
|
Zhang Q, Jiang J. Regulation of Hedgehog Signal Transduction by Ubiquitination and Deubiquitination. Int J Mol Sci 2021; 22:ijms222413338. [PMID: 34948134 PMCID: PMC8703657 DOI: 10.3390/ijms222413338] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/23/2022] Open
Abstract
The Hedgehog (Hh) family of secreted proteins governs embryonic development and adult tissue homeostasis in species ranging from insects to mammals. Deregulation of Hh pathway activity has been implicated in a wide range of human disorders, including congenital diseases and cancer. Hh exerts its biological influence through a conserved signaling pathway. Binding of Hh to its receptor Patched (Ptc), a twelve-span transmembrane protein, leads to activation of an atypical GPCR family protein and Hh signal transducer Smoothened (Smo), which then signals downstream to activate the latent Cubitus interruptus (Ci)/Gli family of transcription factors. Hh signal transduction is regulated by ubiquitination and deubiquitination at multiple steps along the pathway including regulation of Ptc, Smo and Ci/Gli proteins. Here we review the effect of ubiquitination and deubiquitination on the function of individual Hh pathway components, the E3 ubiquitin ligases and deubiquitinases involved, how ubiquitination and deubiquitination are regulated, and whether the underlying mechanisms are conserved from Drosophila to mammals.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing 210061, China
- Correspondence: (Q.Z.); (J.J.)
| | - Jin Jiang
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (Q.Z.); (J.J.)
| |
Collapse
|
12
|
Liu M, Su Y, Peng J, Zhu AJ. Protein modifications in Hedgehog signaling: Cross talk and feedback regulation confer divergent Hedgehog signaling activity. Bioessays 2021; 43:e2100153. [PMID: 34738654 DOI: 10.1002/bies.202100153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
The complexity of the Hedgehog (Hh) signaling cascade has increased over the course of evolution; however, it does not suffice to accommodate the dynamic yet robust requirements of differential Hh signaling activity needed for embryonic development and adult homeostatic maintenance. One solution to solve this dilemma is to apply multiple forms of post-translational modifications (PTMs) to the core Hh signaling components, modulating their abundance, localization, and signaling activity. This review summarizes various forms of protein modifications utilized to regulate Hh signaling, with a special emphasis on crosstalk between different forms of PTMs and their feedback regulation by Hh signaling.
Collapse
Affiliation(s)
- Min Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ying Su
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Jingyu Peng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Alan Jian Zhu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
13
|
Mechanisms of Smoothened Regulation in Hedgehog Signaling. Cells 2021; 10:cells10082138. [PMID: 34440907 PMCID: PMC8391454 DOI: 10.3390/cells10082138] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/05/2021] [Accepted: 08/16/2021] [Indexed: 12/21/2022] Open
Abstract
The seven-transmembrane protein, Smoothened (SMO), has shown to be critical for the hedgehog (HH) signal transduction on the cell membrane (and the cilium in vertebrates). SMO is subjected to multiple types of post-translational regulations, including phosphorylation, ubiquitination, and sumoylation, which alter SMO intracellular trafficking and cell surface accumulation. Recently, SMO is also shown to be regulated by small molecules, such as oxysterol, cholesterol, and phospholipid. The activity of SMO must be very well balanced by these different mechanisms in vivo because the malfunction of SMO will not only cause developmental defects in early stages, but also induce cancers in late stages. Here, we discuss the activation and inactivation of SMO by different mechanisms to better understand how SMO is regulated by the graded HH signaling activity that eventually governs distinct development outcomes.
Collapse
|
14
|
Kim JH, Hanlon CD, Vohra S, Devreotes PN, Andrew DJ. Hedgehog signaling and Tre1 regulate actin dynamics through PI(4,5)P 2 to direct migration of Drosophila embryonic germ cells. Cell Rep 2021; 34:108799. [PMID: 33657369 PMCID: PMC8023404 DOI: 10.1016/j.celrep.2021.108799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 12/21/2020] [Accepted: 02/05/2021] [Indexed: 01/09/2023] Open
Abstract
The Tre1 G-protein coupled receptor (GPCR) was discovered to be required for Drosophila germ cell (GC) coalescence almost two decades ago, yet the molecular events both upstream and downstream of Tre1 activation remain poorly understood. To gain insight into these events, we describe a bona fide null allele and both untagged and tagged versions of Tre1. We find that the primary defect with complete Tre1 loss is the failure of GCs to properly navigate, with GC mis-migration occurring from early stages. We find that Tre1 localizes with F-actin at the migration front, along with PI(4,5)P2; dPIP5K, an enzyme that generates PI(4,5)P2; and dWIP, a protein that binds activated Wiskott-Aldrich syndrome protein (WASP), which stimulates F-actin polymerization. We show that Tre1 is required for polarized accumulation of F-actin, PI(4,5)P2, and dPIP5K. Smoothened also localizes with F-actin at the migration front, and Hh, through Smo, increases levels of Tre1 at the plasma membrane and Tre1’s association with dPIP5K. Kim et al. uncover molecular and cellular events upstream and downstream of the Tre1 G-protein coupled receptor (GPCR), which is required for germ cell navigation in Drosophila. Hedgehog signaling through Smoothened localizes Tre1 to activate F-actin assembly through dPIP5K, PI(4,5)P2, and WASP.
Collapse
Affiliation(s)
- Ji Hoon Kim
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Caitlin D Hanlon
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sunaina Vohra
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter N Devreotes
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Deborah J Andrew
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Liu M, Liu A, Wang J, Zhang Y, Li Y, Su Y, Zhu AJ. Competition between two phosphatases fine-tunes Hedgehog signaling. J Cell Biol 2020; 220:211641. [PMID: 33373452 PMCID: PMC7774589 DOI: 10.1083/jcb.202010078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/26/2020] [Accepted: 12/02/2020] [Indexed: 12/29/2022] Open
Abstract
Hedgehog (Hh) signaling is essential for embryonic development and adult homeostasis. How its signaling activity is fine-tuned in response to fluctuated Hh gradient is less known. Here, we identify protein phosphatase V (PpV), the catalytic subunit of protein phosphatase 6, as a homeostatic regulator of Hh signaling. PpV is genetically upstream of widerborst (wdb), which encodes a regulatory subunit of PP2A that modulates high-level Hh signaling. We show that PpV negatively regulates Wdb stability independent of phosphatase activity of PpV, by competing with the catalytic subunit of PP2A for Wdb association, leading to Wdb ubiquitination and subsequent proteasomal degradation. Thus, regulated Wdb stability, maintained through competition between two closely related phosphatases, ensures graded Hh signaling. Interestingly, PpV expression is regulated by Hh signaling. Therefore, PpV functions as a Hh activity sensor that regulates Wdb-mediated PP2A activity through feedback mechanisms to maintain Hh signaling homeostasis.
Collapse
Affiliation(s)
- Min Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Aiguo Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jie Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Yansong Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Yajuan Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Ying Su
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China,Correspondence to Ying Su:
| | - Alan Jian Zhu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China,Alan Jian Zhu:
| |
Collapse
|
16
|
Bosakova M, Abraham SP, Nita A, Hruba E, Buchtova M, Taylor SP, Duran I, Martin J, Svozilova K, Barta T, Varecha M, Balek L, Kohoutek J, Radaszkiewicz T, Pusapati GV, Bryja V, Rush ET, Thiffault I, Nickerson DA, Bamshad MJ, Rohatgi R, Cohn DH, Krakow D, Krejci P. Mutations in GRK2 cause Jeune syndrome by impairing Hedgehog and canonical Wnt signaling. EMBO Mol Med 2020; 12:e11739. [PMID: 33200460 PMCID: PMC7645380 DOI: 10.15252/emmm.201911739] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
Mutations in genes affecting primary cilia cause ciliopathies, a diverse group of disorders often affecting skeletal development. This includes Jeune syndrome or asphyxiating thoracic dystrophy (ATD), an autosomal recessive skeletal disorder. Unraveling the responsible molecular pathology helps illuminate mechanisms responsible for functional primary cilia. We identified two families with ATD caused by loss-of-function mutations in the gene encoding adrenergic receptor kinase 1 (ADRBK1 or GRK2). GRK2 cells from an affected individual homozygous for the p.R158* mutation resulted in loss of GRK2, and disrupted chondrocyte growth and differentiation in the cartilage growth plate. GRK2 null cells displayed normal cilia morphology, yet loss of GRK2 compromised cilia-based signaling of Hedgehog (Hh) pathway. Canonical Wnt signaling was also impaired, manifested as a failure to respond to Wnt ligand due to impaired phosphorylation of the Wnt co-receptor LRP6. We have identified GRK2 as an essential regulator of skeletogenesis and demonstrate how both Hh and Wnt signaling mechanistically contribute to skeletal ciliopathies.
Collapse
Affiliation(s)
- Michaela Bosakova
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Sara P Abraham
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Alexandru Nita
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Eva Hruba
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Marcela Buchtova
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - S Paige Taylor
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Ivan Duran
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Jorge Martin
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Katerina Svozilova
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| | - Tomas Barta
- Department of Histology and EmbryologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Miroslav Varecha
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Lukas Balek
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
| | | | - Tomasz Radaszkiewicz
- Institute of Experimental BiologyFaculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Ganesh V Pusapati
- Department of BiochemistryStanford UniversityPalo AltoCAUSA
- Department of MedicineStanford UniversityPalo AltoCAUSA
| | - Vitezslav Bryja
- Institute of Experimental BiologyFaculty of ScienceMasaryk UniversityBrnoCzech Republic
| | - Eric T Rush
- Children's Mercy Kansas City, Center for Pediatric Genomic MedicineKansas CityMOUSA
- Department of PediatricsUniversity of MissouriKansas CityMOUSA
| | - Isabelle Thiffault
- Children's Mercy Kansas City, Center for Pediatric Genomic MedicineKansas CityMOUSA
- Department of PediatricsUniversity of MissouriKansas CityMOUSA
| | | | - Michael J Bamshad
- Department of Genome SciencesUniversity of WashingtonSeattleWAUSA
- Department of PediatricsUniversity of WashingtonSeattleWAUSA
- Division of Genetic MedicineSeattle Children's HospitalSeattleWAUSA
| | | | - Rajat Rohatgi
- Department of BiochemistryStanford UniversityPalo AltoCAUSA
- Department of MedicineStanford UniversityPalo AltoCAUSA
| | - Daniel H Cohn
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Molecular Cell and Developmental BiologyUniversity of California at Los AngelesLos AngelesCAUSA
| | - Deborah Krakow
- Department of Orthopaedic SurgeryDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Human GeneticsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Obstetrics and GynecologyDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - Pavel Krejci
- Department of BiologyFaculty of MedicineMasaryk UniversityBrnoCzech Republic
- International Clinical Research CenterSt. Anne's University HospitalBrnoCzech Republic
- Institute of Animal Physiology and Genetics of the CASBrnoCzech Republic
| |
Collapse
|
17
|
Li H, Wang W, Zhang W, Wu G. Structural insight into the recognition between Sufu and fused in the Hedgehog signal transduction pathway. J Struct Biol 2020; 212:107614. [PMID: 32911070 DOI: 10.1016/j.jsb.2020.107614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/15/2020] [Accepted: 09/02/2020] [Indexed: 10/23/2022]
Abstract
Hedgehog signaling plays a crucial role in embryogenesis and adult tissue homeostasis, and mutations of its key components such as Suppressor of fused (Sufu) are closely associated with human diseases. The Ser/Thr kinase Fused (Fu) promotes Hedgehog signaling by phosphorylating the Cubitus interruptus (Ci)/Glioma-associated oncogene homologue (Gli) family of transcription factors. Sufu associates with both Fu and Ci/Gli, but the recognition mechanism between Sufu and Fu remains obscure. Here, our structure of the N-terminal domain (NTD) of Drosophila Sufu (dSufu) in complex with the Sufu-binding site (SBS) of Fu reveals that both main-chain β sheet formation and side-chain hydrophobic interactions contribute to the recognition between Sufu and Fu, and point mutations of highly conserved interface residues eliminated their association. Structural comparison suggests that Fu and Ci/Gli bind on opposite sides of dSufu-NTD, allowing the formation of a Fu-dSufu-Ci ternary complex which facilitates the phosphorylation of Ci/Gli by Fu. Hence, our results provide insights into the Sufu-Fu recognition mechanism.
Collapse
Affiliation(s)
- Hua Li
- State Key Laboratory of Microbial Metabolism, School of Life Science and Biotechnology, The Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Wang
- State Key Laboratory of Microbial Metabolism, School of Life Science and Biotechnology, The Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Science and Biotechnology, The Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Geng Wu
- State Key Laboratory of Microbial Metabolism, School of Life Science and Biotechnology, The Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
18
|
Wang J, Dahmann C. Establishing compartment boundaries in Drosophila wing imaginal discs: An interplay between selector genes, signaling pathways and cell mechanics. Semin Cell Dev Biol 2020; 107:161-169. [PMID: 32732129 DOI: 10.1016/j.semcdb.2020.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 01/02/2023]
Abstract
The partitioning of cells into groups or 'compartments' separated by straight and sharp boundaries is important for tissue formation in animal development. Cells from neighboring compartments are characterized by distinct fates and functions and their continuous separation at compartment boundaries maintains proper tissue organization. Signaling across compartment boundaries can induce the local expression of morphogens that in turn direct growth and patterning of the surrounding cells. Compartment boundaries play therefore an important role in tissue development. Compartment boundaries were first identified in the early 1970s in the Drosophila wing. Here, we review the role of compartment boundaries in growth and patterning of the developing wing and then discuss the genetic and physical mechanisms underlying cell separation at compartment boundaries in this tissue.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
| | - Christian Dahmann
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
19
|
Jiang Y, Zhuo X, Mao C. G Protein-coupled Receptors in Cancer Stem Cells. Curr Pharm Des 2020; 26:1952-1963. [DOI: 10.2174/1381612826666200305130009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022]
Abstract
G protein-coupled receptors (GPCRs) are highly expressed on a variety of tumour tissues while several
GPCR exogenous ligands become marketed pharmaceuticals. In recent decades, cancer stem cells (CSCs) become
widely investigated drug targets for cancer therapy but the underlying mechanism is still not fully elucidated.
There are vigorous participations of GPCRs in CSCs-related signalling and functions, such as biomarkers for
CSCs, activation of Wnt, Hedgehog (HH) and other signalling to facilitate CSCs progressions. This relationship
can not only uncover a novel molecular mechanism for GPCR-mediated cancer cell functions but also assist our
understanding of maintaining and modulating CSCs. Moreover, GPCR antagonists and monoclonal antibodies
could be applied to impair CSCs functions and consequently attenuate tumour growth, some of which have been
undergoing clinical studies and are anticipated to turn into marketed anticancer drugs. Therefore, this review
summarizes and provides sufficient evidences on the regulation of GPCR signalling in the maintenance, differentiation
and pluripotency of CSCs, suggesting that targeting GPCRs on the surface of CSCs could be potential
therapeutic strategies for cancer therapy.
Collapse
Affiliation(s)
- Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xin Zhuo
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Canquan Mao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
20
|
He X, Yan L, Wu Q, Zhang G, Zhou N. Ligand-dependent internalization of Bombyx mori tachykinin-related peptide receptor is regulated by PKC, GRK5 and β-arrestin2/BmKurtz. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118690. [PMID: 32112783 DOI: 10.1016/j.bbamcr.2020.118690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 02/18/2020] [Accepted: 02/23/2020] [Indexed: 10/24/2022]
Abstract
Tachykinin signaling system is present in both vertebrates and invertebrates, and functions as neuromodulator responsible for the regulation of various physiological processes. In human, the internalization of G protein-coupled receptors has been extensively characterized; however, the insect GPCR internalization has been rarely investigated. Here, we constructed two expression vectors of Bombyx tachykinin-related peptide receptor (BmTKRPR) fused with Enhanced Green Fluorescent Protein (EGFP) at the C-terminal end for direct visualization of receptor expression, localization, and trafficking in cultured mammalian HEK293 and insect Sf21 cells. Our results demonstrated that agonist-activated BmTKRPR underwent rapid internalization in a dose-and time-dependent manner via a clathrin-dependent pathway in both HEK293 and Sf21 cells. Further investigation via RNAi or specific inhibitors, or co-immunoprecipitation demonstrated that agonist-induced BmTKRPR internalization was mediated by PKC, GRK5 and β-arrestin2/BmKurtz. In addition, we also observed that most of the internalized BmTKRP receptors were recycled to the cell surface via early endosomes upon peptide ligand removal. Our study provides the first in-depth information on mechanisms underlying insect TKRP receptor internalization and perhaps aids in the interpretation of the signaling in the regulation of physiological processes.
Collapse
Affiliation(s)
- Xiaobai He
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China; College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China.
| | - Lili Yan
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qi Wu
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Guozheng Zhang
- Key Laboratory of Genetic Improvement of Sericulture, Ministry of Agriculture and Rural Affairs, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
21
|
Yang C, Zheng X, Ye K, Sun Y, Lu Y, Fan Q, Ge H. miR-135a Inhibits the Invasion and Migration of Esophageal Cancer Stem Cells through the Hedgehog Signaling Pathway by Targeting Smo. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 19:841-852. [PMID: 31981861 PMCID: PMC6976902 DOI: 10.1016/j.omtn.2019.10.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/22/2019] [Accepted: 10/27/2019] [Indexed: 02/08/2023]
Abstract
Cancer stem cells (CSCs) have been reported to be involved in esophageal cancer (EC) development. Hence, we aim to explore whether microRNA-135a (miR-135a) affects EC and its associated mechanism. Cancerous and adjacent tissues from 138 EC patients were collected. The dual-luciferase reporter gene assay and bioinformatics analysis were used to confirm the interaction between nucleotides. A series of mimics or inhibitors of miR-135a or small interfering RNA (siRNA) against Smo were introduced into EC cells. After that, the expression of miR-135a and Hedgehog (Hh) signaling pathway-related genes (Smo, Gli1, Shh, and Gli2) in tissues and cells was measured, accompanied by evaluation of cell viability, apoptosis, invasion, and migration. High expression of Smo, Gli1, Shh, and Gli2 and low expression of miR-135a were observed in EC. Smo was verified to be a target gene of miR-135a. In addition, overexpression of miR-135a or silencing of Smo decreased the expression of Gli1, Gli2, and Shh, thus inhibiting EC cell proliferation, migration, and invasion and promoting apoptosis. Silencing of miR-135a was observed to reverse the inhibitory role of miR-135a in EC. These results suggest that miR-135a inhibited the migration and invasion of EC cells through inhibition of the Smo/Hh axis.
Collapse
Affiliation(s)
- Chengliang Yang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, P.R. China; Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China
| | - Xiaoli Zheng
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, P.R. China
| | - Ke Ye
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, P.R. China
| | - Yanan Sun
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, P.R. China
| | - Yufei Lu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, P.R. China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P.R. China.
| | - Hong Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, P.R. China.
| |
Collapse
|
22
|
Pathania AS, Ren X, Mahdi MY, Shackleford GM, Erdreich-Epstein A. GRK2 promotes growth of medulloblastoma cells and protects them from chemotherapy-induced apoptosis. Sci Rep 2019; 9:13902. [PMID: 31554835 PMCID: PMC6761358 DOI: 10.1038/s41598-019-50157-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
G-protein coupled receptor kinase 2 (GRK2; ADRBK1, BARK1) is most known as a regulator of G-protein coupled receptors. However, GRK2 also has other functions. Medulloblastomas are the most common malignant brain cancers in children. GRK2 has not been implicated in medulloblastoma biology. Here we report that GRK2 knockdown slowed cell growth, diminished proliferation, and enhanced cisplatin- and etoposide-induced apoptosis in medulloblastoma cell lines UW228-2 and Daoy. Reciprocally, GRK2 overexpression attenuated apoptosis induced by these chemotherapy drugs. Cisplatin and etoposide increased phosphorylation of AKT (S473) and GRK2 knockdown mitigated this increase. Cisplatin and etoposide attenuated ERK phosphorylation, but GRK2 knockdown did not alter this effect. Wildtype GRK2 reversed the increase in cisplatin- and etoposide-induced apoptosis caused by GRK2 knockdown. GRK2-K220R (kinase dead) and GRK2-S670A (unphosphorylated, constitutively active) conferred protection from cisplatin that was similar to wildtype GRK2, suggesting that this protection may be mediated though a kinase-independent activity of GRK2. These data demonstrate that GRK2 contributes to proliferation and survival of these medulloblastoma cell lines and to their protection from cisplatin- and etoposide-induced apoptosis.
Collapse
Affiliation(s)
- Anup S Pathania
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, The Saban Research Institute at Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Xiuhai Ren
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Min Y Mahdi
- Department of Radiology, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Gregory M Shackleford
- Department of Radiology, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Anat Erdreich-Epstein
- Department of Pediatrics, Division of Hematology, Oncology and Blood and Marrow Transplantation, The Saban Research Institute at Children's Hospital Los Angeles and Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA.
- Department of Pathology, Children's Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
23
|
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: 9.4] [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
|
24
|
Han Y, Wang B, Cho YS, Zhu J, Wu J, Chen Y, Jiang J. Phosphorylation of Ci/Gli by Fused Family Kinases Promotes Hedgehog Signaling. Dev Cell 2019; 50:610-626.e4. [PMID: 31279575 DOI: 10.1016/j.devcel.2019.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 04/12/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
Abstract
Hedgehog (Hh) signaling culminates in the conversion of the latent transcription factor Cubitus interruptus (Ci)/Gli into its activator form (CiA/GliA), but the underlying mechanism remains poorly understood. Here, we demonstrate that Hh stimulates the phosphorylation of Ci by the Ser/Thr kinase Fused (Fu) and that Fu-mediated phosphorylation of Ci promotes its activation. We find that Fu directly phosphorylates Ci on Ser218 and Ser1230, which primes its further phosphorylation by CK1 on adjacent sties. These phosphorylation events alter Ci binding to the pathway inhibitor Suppressor of fused (Sufu) and facilitate the recruitment of Transportion and the transcriptional coactivator CBP. Furthermore, we provide evidence that Sonic hedgehog (Shh) activates Gli2 by stimulating its phosphorylation on conserved sites through the Fu-family kinases ULK3 and mFu/STK36 in a manner depending on Gli2 ciliary localization. Hence, Fu-family kinase-mediated phosphorylation of Ci/Gli serves as a conserved mechanism that activates the Hh pathway transcription factor.
Collapse
Affiliation(s)
- Yuhong Han
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Bing Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Yong Suk Cho
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Jian Zhu
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Laboratory of Molecular Oncology, School of Laboratory Medicine, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Henan Province, Xinxiang 453003, China
| | - Jiang Wu
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiaochang Donglu, Yunnan, Kunming 650223, China
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA.
| |
Collapse
|
25
|
Abstract
Signaling pathways that mediate cell-cell communication are essential for collective cell behaviors in multicellular systems. The hedgehog (HH) pathway, first discovered and elucidated in Drosophila, is one of these iconic signaling systems that plays many roles during embryogenesis and in adults; abnormal HH signaling can lead to birth defects and cancer. We review recent structural and biochemical studies that have advanced our understanding of the vertebrate HH pathway, focusing on the mechanisms by which the HH signal is received by patched on target cells, transduced across the cell membrane by smoothened, and transmitted to the nucleus by GLI proteins to influence gene-expression programs.
Collapse
Affiliation(s)
- Jennifer H Kong
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
26
|
Montagnani V, Stecca B. Role of Protein Kinases in Hedgehog Pathway Control and Implications for Cancer Therapy. Cancers (Basel) 2019; 11:cancers11040449. [PMID: 30934935 PMCID: PMC6520855 DOI: 10.3390/cancers11040449] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023] Open
Abstract
Hedgehog (HH) signaling is an evolutionarily conserved pathway that is crucial for growth and tissue patterning during embryonic development. It is mostly quiescent in the adult, where it regulates tissue homeostasis and stem cell behavior. Aberrant reactivation of HH signaling has been associated to several types of cancer, including those in the skin, brain, prostate, breast and hematological malignancies. Activation of the canonical HH signaling is triggered by binding of HH ligand to the twelve-transmembrane protein PATCHED. The binding releases the inhibition of the seven-transmembrane protein SMOOTHENED (SMO), leading to its phosphorylation and activation. Hence, SMO activates the transcriptional effectors of the HH signaling, that belong to the GLI family of transcription factors, acting through a not completely elucidated intracellular signaling cascade. Work from the last few years has shown that protein kinases phosphorylate several core components of the HH signaling, including SMO and the three GLI proteins, acting as powerful regulatory mechanisms to fine tune HH signaling activities. In this review, we will focus on the mechanistic influence of protein kinases on HH signaling transduction. We will also discuss the functional consequences of this regulation and the possible implications for cancer therapy.
Collapse
Affiliation(s)
- Valentina Montagnani
- Core Research Laboratory⁻Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy.
| | - Barbara Stecca
- Core Research Laboratory⁻Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy.
| |
Collapse
|
27
|
Zhang X, Feng L, Qiao N, Liu Y, Zhang DC, Yin H. Cloning, expression pattern and functional characterization of fused, an important kinase of the Hedgehog signalling pathway from Locusta migratoria(Orthoptera: Acridoidea). BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1637781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Xiaohong Zhang
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Li Feng
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Ning Qiao
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Yachao Liu
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Dao Chuan Zhang
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| | - Hong Yin
- College of Life Sciences and the Key Laboratory of Zoological Systematics and Application, Hebei University, Baoding, Hebei, P. R. China
| |
Collapse
|
28
|
Giordano C, Ruel L, Poux C, Therond P. Protein association changes in the Hedgehog signaling complex mediate differential signaling strength. Development 2018; 145:145/24/dev166850. [PMID: 30541874 DOI: 10.1242/dev.166850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 11/07/2018] [Indexed: 01/20/2023]
Abstract
Hedgehog (Hh) is a conserved morphogen that controls cell differentiation and tissue patterning in metazoans. In Drosophila, the Hh signal is transduced from the G protein-coupled receptor Smoothened (Smo) to the cytoplasmic Hh signaling complex (HSC). How activated Smo is translated into a graded activation of the downstream pathway is still not well understood. In this study, we show that the last amino acids of the cytoplasmic tail of Smo, in combination with G protein-coupled receptor kinase 2 (Gprk2), bind to the regulatory domain of Fused (Fu) and highly activate its kinase activity. We further show that this binding induces changes in the association of Fu protein with the HSC and increases the proximity of the Fu catalytic domain to its substrate, the Costal2 kinesin. We propose a new model in which, depending on the magnitude of Hh signaling, Smo and Gprk2 modulate protein association and conformational changes in the HSC, which are responsible for the differential activation of the pathway.
Collapse
Affiliation(s)
- Cecile Giordano
- Université Côte d'Azur, CNRS, Inserm, iBV, 06108 Nice, France
| | - Laurent Ruel
- Université Côte d'Azur, CNRS, Inserm, iBV, 06108 Nice, France
| | - Candice Poux
- Stockholms Universitet, Wenner-Grens Institut, SE-106 91 Stockholm, Sweden
| | - Pascal Therond
- Université Côte d'Azur, CNRS, Inserm, iBV, 06108 Nice, France
| |
Collapse
|
29
|
Liu A. Proteostasis in the Hedgehog signaling pathway. Semin Cell Dev Biol 2018; 93:153-163. [PMID: 31429406 DOI: 10.1016/j.semcdb.2018.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/11/2018] [Accepted: 10/22/2018] [Indexed: 12/29/2022]
Abstract
The Hedgehog (Hh) signaling pathway is crucial for the development of vertebrate and invertebrate animals alike. Hh ligand binds its receptor Patched (Ptc), allowing the activation of the obligate signal transducer Smoothened (Smo). The levels and localizations of both Ptc and Smo are regulated by ubiquitination, and Smo is under additional regulation by phosphorylation and SUMOylation. Downstream of Smo, the Ci/Gli family of transcription factors regulates the transcriptional responses to Hh. Phosphorylation, ubiquitination and SUMOylation are important for the stability and localization of Ci/Gli proteins and Hh signaling output. Finally, Suppressor of Fused directly regulates Ci/Gli proteins and itself is under proteolytic regulation that is critical for normal Hh signaling.
Collapse
Affiliation(s)
- Aimin Liu
- Department of Biology, Eberly College of Science, Huck Institute of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, United States.
| |
Collapse
|
30
|
Li S, Cho YS, Wang B, Li S, Jiang J. Regulation of Smoothened ubiquitylation and cell surface expression through a Cul4-DDB1-Gβ E3 ubiquitin ligase complex. J Cell Sci 2018; 131:jcs.218016. [PMID: 29930086 DOI: 10.1242/jcs.218016] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/15/2018] [Indexed: 12/21/2022] Open
Abstract
Hedgehog (Hh) transduces signals by promoting cell surface accumulation and activation of the G-protein-coupled receptor (GPCR)-family protein Smoothened (Smo) in Drosophila, but the molecular mechanism underlying the regulation of Smo trafficking remains poorly understood. Here, we identified the Cul4-DDB1 E3 ubiquitin ligase complex as being essential for Smo ubiquitylation and cell surface clearance. We found that the C-terminal intracellular domain of Smo recruits Cul4-DDB1 through the β subunit of trimeric G protein (Gβ), and that Cul4-DDB1-Gβ promotes the ubiquitylation of both Smo and its binding partner G-protein-coupled-receptor kinase 2 (Gprk2) and induces the internalization and degradation of Smo. Hh dissociates Cul4-DDB1 from Smo by recruiting the catalytic subunit of protein kinase A (PKA) to phosphorylate DDB1, which disrupts its interaction with Gβ. Inactivation of the Cul4-DDB1 complex resulted in elevated Smo cell surface expression, whereas an excessive amount of Cul4-DDB1 blocked Smo accumulation and attenuated Hh pathway activation. Taken together, our study identifies an E3 ubiquitin ligase complex targeting Smo for ubiquitylation and provides new insight into how Hh signaling regulates Smo trafficking and cell surface expression.
Collapse
Affiliation(s)
- Shuang Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Yong Suk Cho
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Bing Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Shuangxi Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA .,Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| |
Collapse
|
31
|
Praktiknjo SD, Saad F, Maier D, Ip P, Hipfner DR. Activation of Smoothened in the Hedgehog pathway unexpectedly increases Gα s-dependent cAMP levels in Drosophila. J Biol Chem 2018; 293:13496-13508. [PMID: 30018136 DOI: 10.1074/jbc.ra118.001953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 07/13/2018] [Indexed: 12/28/2022] Open
Abstract
Hedgehog (Hh) signaling plays a key role in the development and maintenance of animal tissues. This signaling is mediated by the atypical G protein-coupled receptor (GPCR) Smoothened (Smo). Smo activation leads to signaling through several well-characterized effectors to activate Hh target gene expression. Recent studies have implicated activation of the heterotrimeric G protein subunit Gαi and the subsequent decrease in cellular cAMP levels in promoting the Hh response in flies and mammals. Although Hh stimulation decreases cAMP levels in some insect cell lines, here using a bioluminescence resonance energy transfer (BRET)-based assay we found that this stimulation had no detectable effect in Drosophila S2-R+ cells. However, we observed an unexpected and significant Gαs-dependent increase in cAMP levels in response to strong Smo activation in Smo-transfected cells. This effect was mediated by Smo's broadly conserved core, and was specifically activated in response to phosphorylation of the Smo C-terminus by GPCR kinase 2 (Gprk2). Genetic analysis of heterotrimeric G protein function in the developing Drosophila wing revealed a positive role for cAMP in the endogenous Hh response. Specifically, we found that mutation or depletion of Gαs diminished low-threshold Hh responses in Drosophila, whereas depletion of Gαi potentiated them (in contrast to previous findings). Our analysis suggested that regulated cAMP production is important for controlling the sensitivity of cellular responses to Hh in Drosophila.
Collapse
Affiliation(s)
- Samantha D Praktiknjo
- From the Institut de recherches cliniques de Montréal, Montreal, Quebec H2W 1R7.,the Departments of Anatomy and Cell Biology and
| | - Farah Saad
- From the Institut de recherches cliniques de Montréal, Montreal, Quebec H2W 1R7.,Biology, McGill University, Montreal, Quebec H3A 0C7, and
| | - Dominic Maier
- From the Institut de recherches cliniques de Montréal, Montreal, Quebec H2W 1R7.,the Departments of Anatomy and Cell Biology and
| | - Pamela Ip
- From the Institut de recherches cliniques de Montréal, Montreal, Quebec H2W 1R7.,the Departments of Anatomy and Cell Biology and
| | - David R Hipfner
- From the Institut de recherches cliniques de Montréal, Montreal, Quebec H2W 1R7, .,the Departments of Anatomy and Cell Biology and.,Biology, McGill University, Montreal, Quebec H3A 0C7, and.,the Département de médecine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
32
|
Pusapati GV, Kong JH, Patel BB, Gouti M, Sagner A, Sircar R, Luchetti G, Ingham PW, Briscoe J, Rohatgi R. G protein-coupled receptors control the sensitivity of cells to the morphogen Sonic Hedgehog. Sci Signal 2018; 11:eaao5749. [PMID: 29438014 PMCID: PMC5828112 DOI: 10.1126/scisignal.aao5749] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The morphogen Sonic Hedgehog (SHH) patterns tissues during development by directing cell fates in a concentration-dependent manner. The SHH signal is transmitted across the membrane of target cells by the heptahelical transmembrane protein Smoothened (SMO), which activates the GLI family of transcription factors through a mechanism that is undefined in vertebrates. Using CRISPR-edited null alleles and small-molecule inhibitors, we systematically analyzed the epistatic interactions between SMO and three proteins implicated in SMO signaling: the heterotrimeric G protein subunit GαS, the G protein-coupled receptor kinase 2 (GRK2), and the GαS-coupled receptor GPR161. Our experiments uncovered a signaling mechanism that modifies the sensitivity of target cells to SHH and consequently changes the shape of the SHH dose-response curve. In both fibroblasts and spinal neural progenitors, the loss of GPR161, previously implicated as an inhibitor of basal SHH signaling, increased the sensitivity of target cells across the entire spectrum of SHH concentrations. Even in cells lacking GPR161, GRK2 was required for SHH signaling, and Gαs, which promotes the activation of protein Kinase A (PKA), antagonized SHH signaling. We propose that the sensitivity of target cells to Hedgehog morphogens, and the consequent effects on gene expression and differentiation outcomes, can be controlled by signals from G protein-coupled receptors that converge on Gαs and PKA.
Collapse
Affiliation(s)
- Ganesh V Pusapati
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer H Kong
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bhaven B Patel
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mina Gouti
- The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Andreas Sagner
- The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Ria Sircar
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Giovanni Luchetti
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 639798, Singapore
- Living Systems Institute, University of Exeter, Exeter EX4 4RJ, UK
| | - James Briscoe
- The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Rajat Rohatgi
- Departments of Medicine and Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
33
|
Li S, Li S, Wang B, Jiang J. Hedgehog reciprocally controls trafficking of Smo and Ptc through the Smurf family of E3 ubiquitin ligases. Sci Signal 2018; 11:11/516/eaan8660. [PMID: 29438012 DOI: 10.1126/scisignal.aan8660] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hedgehog (Hh) induces signaling by promoting the reciprocal trafficking of its receptor Patched (Ptc) and the signal transducer Smoothened (Smo), which is inhibited by Ptc, at the cell surface. We identified Smurf family E3 ubiquitin ligases as essential for Smo ubiquitylation and cell surface clearance and demonstrated that Smurf family members mediate the reciprocal trafficking of Ptc and Smo in Drosophila melanogaster G protein-coupled receptor kinase 2 (Gprk2)-mediated phosphorylation of Smurf promoted Smo ubiquitylation by increasing the recruitment of Smurf to Smo, whereas protein kinase A (PKA)-mediated phosphorylation of Smo caused Smurf to dissociate from Smo, thereby inhibiting Smo ubiquitylation. Smo and Ptc competed for the same pool of Smurf family E3 ubiquitin ligases, and Hh promoted Ptc ubiquitylation and degradation by disrupting the association of Smurf family E3 ubiquitin ligases with Smo and stimulating their binding to Ptc. Our study identifies the E3 ubiquitin ligases that target Smo and provides insight into how Hh regulates the reciprocal trafficking of its receptor and signal transducer.
Collapse
Affiliation(s)
- Shuang Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Shuangxi Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Bing Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA. .,Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Wang XF, Shen Y, Cheng Q, Fu CL, Zhou ZZ, Hirose S, Liu QX. Apontic directly activates hedgehog and cyclin E for proper organ growth and patterning. Sci Rep 2017; 7:12470. [PMID: 28963499 PMCID: PMC5622130 DOI: 10.1038/s41598-017-12766-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/14/2017] [Indexed: 11/09/2022] Open
Abstract
Hedgehog (Hh) signaling pathway and Cyclin E are key players in cell proliferation and organ development. Hyperactivation of hh and cyclin E has been linked to several types of cancer. However, coordination of the expression of hh and cyclin E was not well understood. Here we show that an evolutionarily conserved transcription factor Apontic (Apt) directly activates hh and cyclin E through its binding site in the promoter regions of hh and cyclin E. This Apt-dependent proper expression of hh and cyclin E is required for cell proliferation and development of the Drosophila wing. Furthermore, Fibrinogen silencer-binding protein (FSBP), a mammalian homolog of Apt, also positively regulates Sonic hh (Shh), Desert hh (Dhh), Cyclin E1 (CCNE1) and Cyclin E2 (CCNE2) in cultured human cells, suggesting evolutionary conservation of the mechanism. Apt-mediated expression of hh and cyclin E can direct proliferation of Hh-expressing cells and simultaneous growth, patterning and differentiation of Hh-recipient cells. The discovery of the simultaneous expression of Hh and principal cell-cycle regulator Cyclin E by Apt implicates insight into the mechanism by which deregulated hh and cyclin E promotes tumor formation.
Collapse
Affiliation(s)
- Xian-Feng Wang
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Yang Shen
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Qian Cheng
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Chong-Lei Fu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Zi-Zhang Zhou
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Susumu Hirose
- Department of Developmental Genetics, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan.
| | - Qing-Xin Liu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong, 271018, China.
| |
Collapse
|
35
|
Ding M, Wang X. Antagonism between Hedgehog and Wnt signaling pathways regulates tumorigenicity. Oncol Lett 2017; 14:6327-6333. [PMID: 29391876 DOI: 10.3892/ol.2017.7030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/30/2017] [Indexed: 01/16/2023] Open
Abstract
The crosstalk of multiple cellular signaling pathways is crucial in animal development and tissue homeostasis, and its dysregulation may result in tumor formation and metastasis. The Hedgehog (Hh) and Wnt signaling pathways are both considered to be essential regulators of cell proliferation, differentiation and oncogenesis. Recent studies have indicated that the Hh and Wnt signaling pathways are closely associated and involved in regulating embryogenesis and cellular differentiation. Hh signaling acts upstream of the Wnt signaling pathway, and negative regulates Wnt activity via secreted frizzled-related protein 1 (SFRP1), and the Wnt/β-catenin pathway downregulates Hh activity through glioma-associated oncogene homolog 3 transcriptional regulation. This evidence suggests that the imbalance of Hh and Wnt regulation serves a crucial role in cancer-associated processes. The activation of SFRP1, which inhibits Wnt, has been demonstrated to be an important cross-point between the two signaling pathways. The present study reviews the complex interaction between the Hh and Wnt signaling pathways in embryogenesis and tumorigenicity, and the role of SFRP1 as an important mediator associated with the dysregulation of the Hh and Wnt signaling pathways.
Collapse
Affiliation(s)
- Mei Ding
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
36
|
Zhao L, Wang L, Chi C, Lan W, Su Y. The emerging roles of phosphatases in Hedgehog pathway. Cell Commun Signal 2017; 15:35. [PMID: 28931407 PMCID: PMC5607574 DOI: 10.1186/s12964-017-0191-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/14/2017] [Indexed: 01/12/2023] Open
Abstract
Hedgehog signaling is evolutionarily conserved and plays a pivotal role in cell fate determination, embryonic development, and tissue renewal. As aberrant Hedgehog signaling is tightly associated with a broad range of human diseases, its activities must be precisely controlled. It has been known that several core components of Hedgehog pathway undergo reversible phosphorylations mediated by protein kinases and phosphatases, which acts as an effective regulatory mechanism to modulate Hedgehog signal activities. In contrast to kinases that have been extensively studied in these phosphorylation events, phosphatases were thought to function in an unspecific manner, thus obtained much less emphasis in the past. However, in recent years, increasing evidence has implicated that phosphatases play crucial and specific roles in the context of developmental signaling, including Hedgehog signaling. In this review, we present a summary of current progress on phosphatase studies in Hedgehog pathway, emphasizing the multiple employments of protein serine/threonine phosphatases during the transduction of morphogenic Hedgehog signal in both Drosophila and vertebrate systems, all of which provide insights into the importance of phosphatases in the specific regulation of Hedgehog signaling.
Collapse
Affiliation(s)
- Long Zhao
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Liguo Wang
- Institute of Evolution & Marine Biodiversity, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Chunli Chi
- Institute of Evolution & Marine Biodiversity, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Wenwen Lan
- Institute of Evolution & Marine Biodiversity, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
37
|
Abstract
The conserved Hedgehog signaling pathway is essential for embryonic development and tissue homeostasis and is tightly regulated. In this issue of Developmental Cell, Ma et al. (2016) demonstrate that sumoylation works in parallel with phosphorylation to stabilize Smoothened, antagonizing its ubiquitination and subsequent degradation, thus activating Hh target gene transcription.
Collapse
Affiliation(s)
- Yun Qi
- State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Han Liu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
38
|
Chen P, Zhou Z, Yao X, Pang S, Liu M, Jiang W, Jiang J, Zhang Q. Capping Enzyme mRNA-cap/RNGTT Regulates Hedgehog Pathway Activity by Antagonizing Protein Kinase A. Sci Rep 2017; 7:2891. [PMID: 28588207 PMCID: PMC5460166 DOI: 10.1038/s41598-017-03165-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 04/21/2017] [Indexed: 02/07/2023] Open
Abstract
Hedgehog (Hh) signaling plays a pivotal role in animal development and its deregulation in humans causes birth defects and several types of cancer. Protein Kinase A (PKA) modulates Hh signaling activity through phosphorylating the transcription factor Cubitus interruptus (Ci) and G protein coupled receptor (GPCR) family protein Smoothened (Smo) in Drosophila, but how PKA activity is regulated remains elusive. Here, we identify a novel regulator of the Hh pathway, the capping-enzyme mRNA-cap, which positively regulates Hh signaling activity through modulating PKA activity. We provide genetic and biochemical evidence that mRNA-cap inhibits PKA kinase activity to promote Hh signaling. Interestingly, regulation of Hh signaling by mRNA-cap depends on its cytoplasmic capping-enzyme activity. In addition, we show that the mammalian homolog of mRNA-cap, RNGTT, can replace mRNA-cap to play the same function in the Drosophila Hh pathway and that knockdown of Rngtt in cultured mammalian cells compromised Shh pathway activity, suggesting that RNGTT is functionally conserved. Our study makes an unexpected link between the mRNA capping machinery and the Hh signaling pathway, unveils a new facet of Hh signaling regulation, and reveals a potential drug target for modulating Hh signaling activity.
Collapse
Affiliation(s)
- Ping Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Zizhang Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Xia Yao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Shu Pang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Meijing Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Weirong Jiang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA. .,Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390, USA.
| | - Qing Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing, 210061, China.
| |
Collapse
|
39
|
Abstract
The casein kinase 1 (CK1) family of serine (Ser)/threonine (Thr) protein kinases participates in a myriad of cellular processes including developmental signaling. Hedgehog (Hh) and Wnt pathways are two major and evolutionarily conserved signaling pathways that control embryonic development and adult tissue homeostasis. Deregulation of these pathways leads to many human disorders including birth defects and cancer. Here, I review the role of CK1 in the regulation of Hh and Wnt signal transduction cascades from the membrane reception systems to the transcriptional effectors. In both Hh and Wnt pathways, multiple CK1 family members regulate signal transduction at several levels of the pathways and play either positive or negative roles depending on the signaling status, individual CK1 isoforms involved, and the specific substrates they phosphorylate. A common mechanism underlying the control of CK1-mediated phosphorylation of Hh and Wnt pathway components is the regulation of CK1/substrate interaction within large protein complexes. I will highlight this feature in the context of Hh signaling and draw interesting parallels between the Hh and Wnt pathways.
Collapse
Affiliation(s)
- Jin Jiang
- University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States.
| |
Collapse
|
40
|
Regulation of Smoothened Trafficking and Hedgehog Signaling by the SUMO Pathway. Dev Cell 2016; 39:438-451. [PMID: 27746045 DOI: 10.1016/j.devcel.2016.09.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 06/14/2016] [Accepted: 09/13/2016] [Indexed: 12/21/2022]
Abstract
Hedgehog (Hh) signaling plays a central role in development and diseases. Hh activates its signal transducer and GPCR-family protein Smoothened (Smo) by inducing Smo phosphorylation, but whether Smo is activated through other post-translational modifications remains unexplored. Here we show that sumoylation acts in parallel with phosphorylation to promote Smo cell-surface expression and Hh signaling. We find that Hh stimulates Smo sumoylation by dissociating it from a desumoylation enzyme Ulp1. Sumoylation of Smo in turn recruits a deubiquitinase UBPY/USP8 to antagonize Smo ubiquitination and degradation, leading to its cell-surface accumulation and elevated Hh pathway activity. We also provide evidence that Shh stimulates sumoylation of mammalian Smo (mSmo) and that sumoylation promotes ciliary localization of mSmo and Shh pathway activity. Our findings reveal a conserved mechanism whereby the SUMO pathway promotes Hh signaling by regulating Smo subcellular localization and shed light on how sumoylation regulates membrane protein trafficking.
Collapse
|
41
|
Ramsbottom SA, Pownall ME, Roelink H, Conway SJ. Regulation of Hedgehog Signalling Inside and Outside the Cell. J Dev Biol 2016; 4:23. [PMID: 27547735 PMCID: PMC4990124 DOI: 10.3390/jdb4030023] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The hedgehog (Hh) signalling pathway is conserved throughout metazoans and plays an important regulatory role in both embryonic development and adult homeostasis. Many levels of regulation exist that control the release, reception, and interpretation of the hedgehog signal. The fatty nature of the Shh ligand means that it tends to associate tightly with the cell membrane, and yet it is known to act as a morphogen that diffuses to elicit pattern formation. Heparan sulfate proteoglycans (HSPGs) play a major role in the regulation of Hh distribution outside the cell. Inside the cell, the primary cilium provides an important hub for processing the Hh signal in vertebrates. This review will summarise the current understanding of how the Hh pathway is regulated from ligand production, release, and diffusion, through to signal reception and intracellular transduction.
Collapse
Affiliation(s)
- Simon A. Ramsbottom
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, NE1 3BZ Newcastle upon Tyne, UK
- Correspondence: ; Tel.: +44-(0)191-241-8612
| | | | | | | |
Collapse
|
42
|
Li S, Li S, Han Y, Tong C, Wang B, Chen Y, Jiang J. Regulation of Smoothened Phosphorylation and High-Level Hedgehog Signaling Activity by a Plasma Membrane Associated Kinase. PLoS Biol 2016; 14:e1002481. [PMID: 27280464 PMCID: PMC4900676 DOI: 10.1371/journal.pbio.1002481] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/12/2016] [Indexed: 12/21/2022] Open
Abstract
Hedgehog (Hh) signaling controls embryonic development and adult tissue homeostasis through the G protein coupled receptor (GPCR)-family protein Smoothened (Smo). Upon stimulation, Smo accumulates on the cell surface in Drosophila or primary cilia in vertebrates, which is thought to be essential for its activation and function, but the underlying mechanisms remain poorly understood. Here we show that Hh stimulates the binding of Smo to a plasma membrane-associated kinase Gilgamesh (Gish)/CK1γ and that Gish fine-tunes Hh pathway activity by phosphorylating a Ser/Thr cluster (CL-II) in the juxtamembrane region of Smo carboxyl-terminal intracellular tail (C-tail). We find that CL-II phosphorylation is promoted by protein kinase A (PKA)-mediated phosphorylation of Smo C-tail and depends on cell surface localization of both Gish and Smo. Consistent with CL-II being critical for high-threshold Hh target gene expression, its phosphorylation appears to require higher levels of Hh or longer exposure to the same level of Hh than PKA-site phosphorylation on Smo. Furthermore, we find that vertebrate CK1γ is localized at the primary cilium to promote Smo phosphorylation and Sonic hedgehog (Shh) pathway activation. Our study reveals a conserved mechanism whereby Hh induces a change in Smo subcellular localization to promote its association with and activation by a plasma membrane localized kinase, and provides new insight into how Hh morphogen progressively activates Smo.
Collapse
Affiliation(s)
- Shuangxi Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Shuang Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Yuhong Han
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Chao Tong
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Bing Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Yongbin Chen
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
43
|
Zhao Z, Lee RTH, Pusapati GV, Iyu A, Rohatgi R, Ingham PW. An essential role for Grk2 in Hedgehog signalling downstream of Smoothened. EMBO Rep 2016; 17:739-52. [PMID: 27113758 PMCID: PMC5341524 DOI: 10.15252/embr.201541532] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 03/07/2016] [Indexed: 01/28/2023] Open
Abstract
The G‐protein‐coupled receptor kinase 2 (adrbk2/GRK2) has been implicated in vertebrate Hedgehog (Hh) signalling based on the effects of its transient knock‐down in mammalian cells and zebrafish embryos. Here, we show that the response to Hh signalling is effectively abolished in the absence of Grk2 activity. Zebrafish embryos lacking all Grk2 activity are refractory to both Sonic hedgehog (Shh) and oncogenic Smoothened (Smo) activity, but remain responsive to inhibition of cAMP‐dependent protein kinase (PKA) activity. Mutation of the kinase domain abrogates the rescuing activity of grk2 mRNA, suggesting that Grk2 acts in a kinase‐dependent manner to regulate the response to Hh. Previous studies have suggested that Grk2 potentiates Smo activity by phosphorylating its C‐terminal tail (CTT). In the zebrafish embryo, however, phosphomimetic Smo does not display constitutive activity, whereas phospho‐null mutants retain activity, implying phosphorylation is neither sufficient nor necessary for Smo function. Since Grk2 rescuing activity requires the integrity of domains essential for its interaction with GPCRs, we speculate that Grk2 may regulate Hh pathway activity by downregulation of a GPCR.
Collapse
Affiliation(s)
- Zhonghua Zhao
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore Developmental and Biomedical Genetics Laboratory, Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-STAR), Singapore, Singapore
| | - Raymond Teck Ho Lee
- Developmental and Biomedical Genetics Laboratory, Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-STAR), Singapore, Singapore
| | - Ganesh V Pusapati
- Departments of Medicine and Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Audrey Iyu
- Developmental and Biomedical Genetics Laboratory, Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-STAR), Singapore, Singapore
| | - Rajat Rohatgi
- Departments of Medicine and Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore Developmental and Biomedical Genetics Laboratory, Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-STAR), Singapore, Singapore
| |
Collapse
|
44
|
Jiang K, Liu Y, Fan J, Zhang J, Li XA, Evers BM, Zhu H, Jia J. PI(4)P Promotes Phosphorylation and Conformational Change of Smoothened through Interaction with Its C-terminal Tail. PLoS Biol 2016; 14:e1002375. [PMID: 26863604 PMCID: PMC4749301 DOI: 10.1371/journal.pbio.1002375] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/07/2016] [Indexed: 12/29/2022] Open
Abstract
In Hedgehog (Hh) signaling, binding of Hh to the Patched-Interference Hh (Ptc-Ihog) receptor complex relieves Ptc inhibition on Smoothened (Smo). A longstanding question is how Ptc inhibits Smo and how such inhibition is relieved by Hh stimulation. In this study, we found that Hh elevates production of phosphatidylinositol 4-phosphate (PI(4)P). Increased levels of PI(4)P promote, whereas decreased levels of PI(4)P inhibit, Hh signaling activity. We further found that PI(4)P directly binds Smo through an arginine motif, which then triggers Smo phosphorylation and activation. Moreover, we identified the pleckstrin homology (PH) domain of G protein-coupled receptor kinase 2 (Gprk2) as an essential component for enriching PI(4)P and facilitating Smo activation. PI(4)P also binds mouse Smo (mSmo) and promotes its phosphorylation and ciliary accumulation. Finally, Hh treatment increases the interaction between Smo and PI(4)P but decreases the interaction between Ptc and PI(4)P, indicating that, in addition to promoting PI(4)P production, Hh regulates the pool of PI(4)P associated with Ptc and Smo.
Collapse
Affiliation(s)
- Kai Jiang
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Yajuan Liu
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Junkai Fan
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jie Zhang
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Xiang-An Li
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - B. Mark Evers
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Surgery, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jianhang Jia
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
45
|
Abstract
The Hedgehog (Hh) signaling pathway play critical roles in embryonic development and adult tissue homeostasis. A critical step in Hh signal transduction is how Hh receptor Patched (Ptc) inhibits the atypical G protein-coupled receptor Smoothened (Smo) in the absence of Hh and how this inhibition is release by Hh stimulation. It is unlikely that Ptc inhibits Smo by direct interaction. Here we discuss how Hh regulates the phosphorylation and ubiquitination of Smo, leading to cell surface and ciliary accumulation of Smo in Drosophila and vertebrate cells, respectively. In addition, we discuss how PI(4)P phospholipid acts in between Ptc and Smo to regulate Smo phosphorylation and activation in response to Hh stimulation.
Collapse
Affiliation(s)
- Kai Jiang
- Markey Cancer Center, Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jianhang Jia
- Markey Cancer Center, Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| |
Collapse
|
46
|
Zhang YR, Gui LS, Li YK, Jiang BJ, Wang HC, Zhang YY, Zan LS. Molecular Characterization of Bovine SMO Gene and Effects of Its Genetic Variations on Body Size Traits in Qinchuan Cattle (Bos taurus). Int J Mol Sci 2015. [PMID: 26225956 PMCID: PMC4581179 DOI: 10.3390/ijms160816966] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Smoothened (Smo)-mediated Hedgehog (Hh) signaling pathway governs the patterning, morphogenesis and growth of many different regions within animal body plans. This study evaluated the effects of genetic variations of the bovine SMO gene on economically important body size traits in Chinese Qinchuan cattle. Altogether, eight single nucleotide polymorphisms (SNPs: 1-8) were identified and genotyped via direct sequencing covering most of the coding region and 3'UTR of the bovine SMO gene. Both the p.698Ser.>Ser. synonymous mutation resulted from SNP1 and the p.700Ser.>Pro. non-synonymous mutation caused by SNP2 mapped to the intracellular C-terminal tail of bovine Smo protein; the other six SNPs were non-coding variants located in the 3'UTR. The linkage disequilibrium was analyzed, and five haplotypes were discovered in 520 Qinchuan cattle. Association analyses showed that SNP2, SNP3/5, SNP4 and SNP6/7 were significantly associated with some body size traits (p < 0.05) except SNP1/8 (p > 0.05). Meanwhile, cattle with wild-type combined haplotype Hap1/Hap1 had significantly (p < 0.05) greater body length than those with Hap2/Hap2. Our results indicate that variations in the SMO gene could affect body size traits of Qinchuan cattle, and the wild-type haplotype Hap1 together with the wild-type alleles of these detected SNPs in the SMO gene could be used to breed cattle with superior body size traits. Therefore, our results could be helpful for marker-assisted selection in beef cattle breeding programs.
Collapse
Affiliation(s)
- Ya-Ran Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Lin-Sheng Gui
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Yao-Kun Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Bi-Jie Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Hong-Cheng Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Ying-Ying Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Lin-Sen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
- National Beef Cattle Improvement Center of Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
47
|
Deubiquitination of Ci/Gli by Usp7/HAUSP Regulates Hedgehog Signaling. Dev Cell 2015; 34:58-72. [PMID: 26120032 DOI: 10.1016/j.devcel.2015.05.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 03/18/2015] [Accepted: 05/19/2015] [Indexed: 01/20/2023]
Abstract
Hedgehog (Hh) signaling plays essential roles in animal development and tissue homeostasis, and its misregulation causes congenital diseases and cancers. Regulation of the ubiquitin/proteasome-mediated proteolysis of Ci/Gli transcription factors is central to Hh signaling, but whether deubiquitinase is involved in this process remains unknown. Here, we show that Hh stimulates the binding of a ubiquitin-specific protease Usp7 to Ci, which positively regulates Hh signaling activity through inhibiting Ci ubiquitination and degradation mediated by both Slimb-Cul1 and Hib-Cul3 E3 ligases. Furthermore, we find that Usp7 forms a complex with GMP-synthetase (GMPS) to promote Hh pathway activity. Finally, we show that the mammalian counterpart of Usp7, HAUSP, positively regulates Hh signaling by modulating Gli ubiquitination and stability. Our findings reveal a conserved mechanism by which Ci/Gli is stabilized by a deubiquitination enzyme and identify Usp7/HUASP as a critical regulator of Hh signaling and potential therapeutic target for Hh-related cancers.
Collapse
|
48
|
Jiang K, Jia J. Analysis of Smoothened Phosphorylation and Activation in Cultured Cells and Wing Discs of Drosophila. Methods Mol Biol 2015; 1322:45-60. [PMID: 26179038 DOI: 10.1007/978-1-4939-2772-2_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Smoothened (Smo) is essential for transduction of the Hedgehog (Hh) signal in both insects and vertebrates. Binding of Hh to Ptc-Ihog relieves the Patched (Ptc)-mediated inhibition of Smo, which allows Smo to activate the cubitus interruptus (Ci)/Gli family of zinc finger transcription factors and thereby induce the expression of Hh target genes, such as decapentaplegic (dpp), ptc, and engrailed (en). The activation of Smo appears to be one of the most important events in Hh signaling. Studies have shown that Hh induces cell surface/ciliary accumulation and phosphorylation of Smo by multiple kinases, including protein kinase A (PKA), casein kinase 1 (CK1), casein kinase 2 (CK2), G protein-coupled receptor kinase 2 (Gprk2), and atypical PKC (aPKC). Here, we describe the assays used to examine the activity of Smo in Hh signaling, including in vitro kinase, ptc-luciferase reporter assay, cell surface accumulation assay, fluorescence resonance energy transfer (FRET) assay, and wing disc immunostaining. These assays are powerful tools to study Smo phosphorylation and activation, which have provided mechanistic insight into a better understanding the mechanisms of Smo regulation.
Collapse
Affiliation(s)
- Kai Jiang
- Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, 741 S. Limestone, BBSRB 373, Lexington, KY, 40536-0509, USA
| | | |
Collapse
|
49
|
Hedgehog-induced phosphorylation by CK1 sustains the activity of Ci/Gli activator. Proc Natl Acad Sci U S A 2014; 111:E5651-60. [PMID: 25512501 DOI: 10.1073/pnas.1416652111] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hedgehog (Hh) signaling governs many developmental processes by regulating the balance between the repressor (Ci(R)/Gli(R)) and activator (Ci(A)/Gli(A)) forms of Cubitus interruptus (Ci)/glioma-associated oncogene homolog (Gli) transcription factors. Although much is known about how Ci(R)/Gli(R) is controlled, the regulation of Ci(A)/Gli(A) remains poorly understood. Here we demonstrate that Casein kinase 1 (CK1) sustains Hh signaling downstream of Costal2 and Suppressor of fused (Sufu) by protecting Ci(A) from premature degradation. We show that Hh stimulates Ci phosphorylation by CK1 at multiple Ser/Thr-rich degrons to inhibit its recognition by the Hh-induced MATH and BTB domain containing protein (HIB), a substrate receptor for the Cullin 3 family of E3 ubiquitin ligases. In Hh-receiving cells, reduction of CK1 activity accelerated HIB-mediated degradation of Ci(A), leading to premature loss of pathway activity. We also provide evidence that Gli(A) is regulated by CK1 in a similar fashion and that CK1 acts downstream of Sufu to promote Sonic hedgehog signaling. Taken together, our study not only reveals an unanticipated and conserved mechanism by which phosphorylation of Ci/Gli positively regulates Hh signaling but also provides the first evidence, to our knowledge, that substrate recognition by the Cullin 3 family of E3 ubiquitin ligases is negatively regulated by a kinase.
Collapse
|
50
|
Hedgehog-regulated atypical PKC promotes phosphorylation and activation of Smoothened and Cubitus interruptus in Drosophila. Proc Natl Acad Sci U S A 2014; 111:E4842-50. [PMID: 25349414 DOI: 10.1073/pnas.1417147111] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Smoothened (Smo) is essential for transduction of the Hedgehog (Hh) signal in both insects and vertebrates. Cell surface/cilium accumulation of Smo is thought to play an important role in Hh signaling, but how the localization of Smo is controlled remains poorly understood. In this study, we demonstrate that atypical PKC (aPKC) regulates Smo phosphorylation and basolateral accumulation in Drosophila wings. Inactivation of aPKC by either RNAi or a mutation inhibits Smo basolateral accumulation and attenuates Hh target gene expression. In contrast, expression of constitutively active aPKC elevates basolateral accumulation of Smo and promotes Hh signaling. The aPKC-mediated phosphorylation of Smo at Ser680 promotes Ser683 phosphorylation by casein kinase 1 (CK1), and these phosphorylation events elevate Smo activity in vivo. Moreover, aPKC has an additional positive role in Hh signaling by regulating the activity of Cubitus interruptus (Ci) through phosphorylation of the Zn finger DNA-binding domain. Finally, the expression of aPKC is up-regulated by Hh signaling in a Ci-dependent manner. Our findings indicate a direct involvement of aPKC in Hh signaling beyond its role in cell polarity.
Collapse
|