1
|
Jung JH, Lee H, Jeon J, Lee YJ, Nada H, Kim M, Lee H, Bhattarai D, Lee K, Ko HW. A novel indole derivative, 2-{3-[1-(benzylsulfonyl)piperidin-4-yl]-2-methyl-1H-indol-1-yl}-1-(pyrrolidin-1-yl)ethenone, suppresses hedgehog signaling and drug-resistant tumor growth. Arch Pharm (Weinheim) 2024; 357:e2400218. [PMID: 38963677 DOI: 10.1002/ardp.202400218] [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: 03/25/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024]
Abstract
The Hedgehog (Hh) signaling pathway plays important roles in various physiological functions. Several malignancies, such as basal cell carcinoma (BCC) and medulloblastoma (MB), have been linked to the aberrant activation of Hh signaling. Although therapeutic drugs have been developed to inhibit Hh pathway-dependent cancer growth, drug resistance remains a major obstacle in cancer treatment. Here, we show that the newly identified, 2-{3-[1-(benzylsulfonyl)-1,2,3,6-tetrahydropyridin-4-yl]-2-methyl-1H-indol-1-yl}-1-(pyrrolidin-1-yl)ethenone analog (LKD1214) exhibits comparable potency to vismodegib in suppressing the Hh pathway activation. LKD1214 represses Smoothened (SMO) activity by blocking its ciliary translocation. Interestingly, we also identified that it has a distinctive binding interface with SMO compared with other SMO-regulating chemicals. Notably, it maintains an inhibitory activity against the SmoD477H mutant, as observed in a patient with vismodegib-resistant BCC. Furthermore, LKD1214 inhibits tumor growth in the mouse model of MB. Collectively, these findings suggest that LKD1214 has the therapeutic potential to overcome drug-resistance in Hh-dependent cancers.
Collapse
Affiliation(s)
- Joo Hyun Jung
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- College of Pharmacy, Dongguk University, Goyangsi, Gyeonggido, Korea
| | - Hwayoung Lee
- College of Pharmacy, Dongguk University, Goyangsi, Gyeonggido, Korea
| | - Jiyeon Jeon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Yoon Ji Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- College of Pharmacy, Dongguk University, Goyangsi, Gyeonggido, Korea
| | - Hossam Nada
- College of Pharmacy, Dongguk University, Goyangsi, Gyeonggido, Korea
| | - Minkyoung Kim
- College of Pharmacy, Dongguk University, Goyangsi, Gyeonggido, Korea
| | - Hankyu Lee
- College of Pharmacy, Dongguk University, Goyangsi, Gyeonggido, Korea
| | - Deepak Bhattarai
- College of Pharmacy, Dongguk University, Goyangsi, Gyeonggido, Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University, Goyangsi, Gyeonggido, Korea
| | - Hyuk Wan Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| |
Collapse
|
2
|
Tingey M, Ruba A, Jiang Z, Yang W. Deciphering vesicle-assisted transport mechanisms in cytoplasm to cilium trafficking. Front Cell Neurosci 2024; 18:1379976. [PMID: 38860265 PMCID: PMC11163138 DOI: 10.3389/fncel.2024.1379976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
The cilium, a pivotal organelle crucial for cell signaling and proper cell function, relies on meticulous macromolecular transport from the cytoplasm for its formation and maintenance. While the intraflagellar transport (IFT) pathway has traditionally been the focus of extensive study concerning ciliogenesis and ciliary maintenance, recent research highlights a complementary and alternative mechanism-vesicle-assisted transport (VAT) in cytoplasm to cilium trafficking. Despite its potential significance, the VAT pathway remains largely uncharacterized. This review explores recent studies providing evidence for the dynamics of vesicle-related diffusion and transport within the live primary cilium, employing high-speed super-resolution light microscopy. Additionally, we analyze the spatial distribution of vesicles in the cilium, mainly relying on electron microscopy data. By scrutinizing the VAT pathways that facilitate cargo transport into the cilium, with a specific emphasis on recent advancements and imaging data, our objective is to synthesize a comprehensive model of ciliary transport through the integration of IFT-VAT mechanisms.
Collapse
Affiliation(s)
| | | | | | - Weidong Yang
- Department of Biology, Temple University, Philadelphia, PA, United States
| |
Collapse
|
3
|
Thazhackavayal Baby B, Kulkarni AM, Gayam PKR, Harikumar KB, Aranjani JM. Beyond cyclopamine: Targeting Hedgehog signaling for cancer intervention. Arch Biochem Biophys 2024; 754:109952. [PMID: 38432565 DOI: 10.1016/j.abb.2024.109952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Hedgehog (Hh) signaling plays a significant role in embryogenesis and several physiological processes, such as wound healing and organ homeostasis. In a pathological setting, it is associated with oncogenesis and is responsible for disease progression and poor clinical outcomes. Hedgehog signaling mediates downstream actions via Glioma Associated Oncogene Homolog (GLI) transcription factors. Inhibiting Hh signaling is an important oncological strategy in which inhibitors of the ligands SMO or GLI have been looked at. This review briefly narrates the Hh ligands, signal transduction, the target genes involved and comprehensively describes the numerous inhibitors that have been evaluated for use in various neoplastic settings.
Collapse
Affiliation(s)
- Beena Thazhackavayal Baby
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka, 576104, India
| | - Aniruddha Murahar Kulkarni
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka, 576104, India
| | - Prasanna Kumar Reddy Gayam
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka, 576104, India
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, 695014, Kerala State, India
| | - Jesil Mathew Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka, 576104, India.
| |
Collapse
|
4
|
Carotenuto P, Gradilone SA, Franco B. Cilia and Cancer: From Molecular Genetics to Therapeutic Strategies. Genes (Basel) 2023; 14:1428. [PMID: 37510333 PMCID: PMC10379587 DOI: 10.3390/genes14071428] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
Cilia are microtubule-based organelles that project from the cell surface with motility or sensory functions. Primary cilia work as antennae to sense and transduce extracellular signals. Cilia critically control proliferation by mediating cell-extrinsic signals and by regulating cell cycle entry. Recent studies have shown that primary cilia and their associated proteins also function in autophagy and genome stability, which are important players in oncogenesis. Abnormal functions of primary cilia may contribute to oncogenesis. Indeed, defective cilia can either promote or suppress cancers, depending on the cancer-initiating mutation, and the presence or absence of primary cilia is associated with specific cancer types. Together, these findings suggest that primary cilia play important, but distinct roles in different cancer types, opening up a completely new avenue of research to understand the biology and treatment of cancers. In this review, we discuss the roles of primary cilia in promoting or inhibiting oncogenesis based on the known or predicted functions of cilia and cilia-associated proteins in several key processes and related clinical implications.
Collapse
Affiliation(s)
- Pietro Carotenuto
- Medical Genetics, Department of Translational Medical Science, University of Naples “Federico II”, 80131 Naples, Italy
- TIGEM, Telethon Institute of Genetics and Medicine, 80078 Naples, Italy
| | - Sergio A. Gradilone
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA;
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brunella Franco
- Medical Genetics, Department of Translational Medical Science, University of Naples “Federico II”, 80131 Naples, Italy
- TIGEM, Telethon Institute of Genetics and Medicine, 80078 Naples, Italy
- School of Advanced Studies, Genomic and Experimental medicine Program (Scuola Superiore Meridionale), 80138 Naples, Italy
| |
Collapse
|
5
|
Bagka M, Choi H, Héritier M, Schwaemmle H, Pasquer QTL, Braun SMG, Scapozza L, Wu Y, Hoogendoorn S. Targeted protein degradation reveals BET bromodomains as the cellular target of Hedgehog pathway inhibitor-1. Nat Commun 2023; 14:3893. [PMID: 37393376 PMCID: PMC10314895 DOI: 10.1038/s41467-023-39657-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 06/22/2023] [Indexed: 07/03/2023] Open
Abstract
Target deconvolution of small molecule hits from phenotypic screens presents a major challenge. Many screens have been conducted to find inhibitors for the Hedgehog signaling pathway - a developmental pathway with many implications in health and disease - yielding many hits but only few identified cellular targets. We here present a strategy for target identification based on Proteolysis-Targeting Chimeras (PROTACs), combined with label-free quantitative proteomics. We develop a PROTAC based on Hedgehog Pathway Inhibitor-1 (HPI-1), a phenotypic screen hit with unknown cellular target. Using this Hedgehog Pathway PROTAC (HPP) we identify and validate BET bromodomains as the cellular targets of HPI-1. Furthermore, we find that HPP-9 is a long-acting Hedgehog pathway inhibitor through prolonged BET bromodomain degradation. Collectively, we provide a powerful PROTAC-based approach for target deconvolution, that answers the longstanding question of the cellular target of HPI-1 and yields a PROTAC that acts on the Hedgehog pathway.
Collapse
Affiliation(s)
- Meropi Bagka
- Department of Organic Chemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Hyeonyi Choi
- Department of Organic Chemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Margaux Héritier
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Hanna Schwaemmle
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Quentin T L Pasquer
- Department of Organic Chemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Simon M G Braun
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Yibo Wu
- Chemical Biology Mass Spectrometry Platform (CHEMBIOMS), Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Sascha Hoogendoorn
- Department of Organic Chemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
6
|
A Potent Antagonist of Smoothened in Hedgehog Signaling for Epilepsy. Int J Mol Sci 2022; 23:ijms232314505. [PMID: 36498832 PMCID: PMC9739937 DOI: 10.3390/ijms232314505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Epilepsy is one of the common encephalopathies caused by sudden abnormal discharges of neurons in the brain. About 30% of patients with epilepsy are insensitive and refractory to existing antiseizure medications. The sonic hedgehog signaling pathway is essential to the development and homeostasis of brain. Aberrant sonic hedgehog signaling is increased in refractory epileptic lesions and may involve the etiology of epilepsy. Thus, new inhibitors of Smoothened, a key signal transducer of this signaling pathway are urgently need for refractory epilepsy. We have established a high-throughput screening platform and discovered several active small molecules targeting Smoothened including TT22. Here we show that the novel Smoothened inhibitor TT22 could block the translocation of βarrestin2-GFP to Smoothened, reduce the accumulation of Smoothened on primary cilia, displace Bodipy-cyclopamine binding to Smoothened, and inhibit the expression of downstream Gli transcription factor. Moreover, TT22 inhibits the abnormal seizure-like activity in neurons. Furthermore, we demonstrated that FDA-approved Smoothened inhibitor GDC-0449 and LDE-225 are able to inhibit abnormal seizure-like activity in neurons. Thus, our study suggests that targeting the sonic hedgehog signaling with new small-molecule Smoothened inhibitors might provide a potential new therapeutic avenue for refractory epilepsy.
Collapse
|
7
|
Rajagopalan S, Singh A, Khiabanian H. Cilium Expression Score Predicts Glioma Survival. Front Genet 2021; 12:758391. [PMID: 34868236 PMCID: PMC8640099 DOI: 10.3389/fgene.2021.758391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/02/2021] [Indexed: 01/29/2023] Open
Abstract
The accurate classification, prognostication, and treatment of gliomas has been hindered by an existing cellular, genomic, and transcriptomic heterogeneity within individual tumors and their microenvironments. Traditional clustering is limited in its ability to distinguish heterogeneity in gliomas because the clusters are required to be exclusive and exhaustive. In contrast, biclustering can identify groups of co-regulated genes with respect to a subset of samples and vice versa. In this study, we analyzed 1,798 normal and tumor brain samples using an unsupervised biclustering approach. We identified co-regulated gene expression profiles that were linked to proximally located brain regions and detected upregulated genes in subsets of gliomas, associated with their histologic grade and clinical outcome. In particular, we present a cilium-associated signature that when upregulated in tumors is predictive of poor survival. We also introduce a risk score based on expression of 12 cilium-associated genes which is reproducibly informative of survival independent of other prognostic biomarkers. These results highlight the role of cilia in development and progression of gliomas and suggest potential therapeutic vulnerabilities for these highly aggressive tumors.
Collapse
Affiliation(s)
- Srinivas Rajagopalan
- Center for Systems and Computational Biology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, United States
| | - Amartya Singh
- Center for Systems and Computational Biology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, United States
| | - Hossein Khiabanian
- Center for Systems and Computational Biology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, United States.,Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, United States
| |
Collapse
|
8
|
Wei SF, He DH, Zhang SB, Lu Y, Ye X, Fan XZ, Wang H, Wang Q, Liu YQ. Identification of pseudolaric acid B as a novel Hedgehog pathway inhibitor in medulloblastoma. Biochem Pharmacol 2021; 190:114593. [PMID: 33964282 DOI: 10.1016/j.bcp.2021.114593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 10/21/2022]
Abstract
Aberrant activation of the Hedgehog (Hh) pathway is implicated in the pathogenesis and development of multiple cancers, especially Hh-driven medulloblastoma (MB). Smoothened (SMO) is a promising therapeutic target of the Hh pathway in clinical cancer treatment. However, SMO mutations frequently occur, which leads to drug resistance and tumor relapse. Novel inhibitors that target both the wild-type and mutant SMO are in high demand. In this study, we identified a novel Hh pathway inhibitor, pseudolaric acid B (PAB), which significantly inhibited the expression of Gli1 and its transcriptional target genes, such as cyclin D1 and N-myc, thus inhibiting the proliferation of DAOY and Ptch1+/- primary MB cells. Mechanistically, PAB can potentially bind to the extracellular entrance of the heptahelical transmembrane domain (TMD) of SMO, based on molecular docking and the BODIPY-cyclopamine binding assay. Further, PAB also efficiently blocked ciliogenesis, demonstrating the inhibitory effects of PAB on the Hh pathway at multiple levels. Thus, PAB may overcome drug-resistance induced by SMO mutations, which frequently occurs in clinical setting. PAB markedly suppressed tumor growth in the subcutaneous allografts of Ptch1+/- MB cells. Together, our results identified PAB as a potent Hh pathway inhibitor to treat Hh-dependent MB, especially cases resistant to SMO antagonists.
Collapse
Affiliation(s)
- Su-Fen Wei
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Dan-Hua He
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Research Center of Chinese Herbal Resources Science and Engineering, School of Pharmaceutical Sciences; Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Shi-Bing Zhang
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yongzhi Lu
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China; State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xiaowei Ye
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiang-Zhen Fan
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hong Wang
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Yong-Qiang Liu
- Institute of Clinical Pharmacology, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Research Center of Chinese Herbal Resources Science and Engineering, School of Pharmaceutical Sciences; Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
9
|
Fan J, Li H, Kuang L, Zhao Z, He W, Liu C, Wang Y, Cheng SY, Chen W. Identification of a potent antagonist of smoothened in hedgehog signaling. Cell Biosci 2021; 11:46. [PMID: 33653381 PMCID: PMC7923671 DOI: 10.1186/s13578-021-00558-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/16/2021] [Indexed: 12/31/2022] Open
Abstract
Background Hedgehog signaling is essential to the regulation of embryonic development, tissue homeostasis, and stem cell self-renewal, making it a prime target for developing cancer therapeutics. Given the close link between aberrant Hedgehog signaling and cancers, many small molecular compounds have been developed to inhibit Smoothened, a key signal transducer of this pathway, for treating cancer and several such compounds have been approved by the United States Food and Drug Administration (GDC-0449 and LDE-225). However, acquired drug resistance has emerged as an important obstacle to the effective use of these first generation Hedgehog pathway blockers. Thus, new Smoothened inhibitors that can overcome such resistance is an urgent need going forward. Results We established the Smoothened/βarrestin2-GFP high-throughput screening platform based on the mechanistic discovery of Hedgehog signaling pathway, and discovered several active small molecules targeting Smoothened including 0025A. Here we show that 0025A can block the translocation of βarrestin2-GFP to Smoothened, displace Bodipy-cyclopamine binding to wild-type Smoothened or mutant Smoothened-D473H, reduce the accumulation of Smo on primary cilia and the expression of Gli upon Hedgehog stimulation. In addition, we show that 0025A can effectively suppress hair follicle morphogenesis and hair growth in mice. Conclusions Our results demonstrate that 0025A is a potent antagonist targeting Smoothened wild-type and mutant receptors in the Hedgehog signaling pathway and may provide a new therapy for refractory cancers.
Collapse
Affiliation(s)
- Junwan Fan
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, 100070, China
| | - Haowen Li
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, 100070, China
| | - Lun Kuang
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Zichen Zhao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, 100070, China
| | - Wenyan He
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, 100070, China
| | - Chen Liu
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Yongjun Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China. .,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, 100070, China.
| | - Steven Y Cheng
- Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210000, Jiangsu, China.
| | - Wei Chen
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China. .,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, 100070, China.
| |
Collapse
|
10
|
Bose C, Banerjee P, Kundu J, Dutta B, Ghosh I, Sinha S, Ghosh A, Barua A, Gupta S, Das U, Jana SS, Sinha S. Evaluation of a Tubulin‐Targeted Pyrimidine Indole Hybrid Molecule as an Anticancer Agent. ChemistrySelect 2020. [DOI: 10.1002/slct.202003322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Chandra Bose
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Priyanjalee Banerjee
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Jayanta Kundu
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Biswadeb Dutta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Indranil Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032 India
| | - Shreya Sinha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Argha Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032 India
| | - Abhishek Barua
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032 India
| | - Shalini Gupta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Ujjal Das
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Siddhartha S. Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032 India
| | - Surajit Sinha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| |
Collapse
|
11
|
Wang B, Liang Z, Liu P. Functional aspects of primary cilium in signaling, assembly and microenvironment in cancer. J Cell Physiol 2020; 236:3207-3219. [PMID: 33107052 PMCID: PMC7984063 DOI: 10.1002/jcp.30117] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/16/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022]
Abstract
The primary cilium is an antennae‐like structure extent outside the cell surface. It has an important role in regulating cell‐signaling transduction to affect proliferation, differentiation and migration. Evidence is accumulating that ciliary defects lead to ciliopathies and ciliary deregulation also play crucial roles in cancer formation and progression. Interestingly, restoring the cilia can suppress proliferation in some cancer cell. However, t he role of primary cilia in cancer still be debated. In this article, we review the role of the primary cilium in cancer through architecture, signaling pathways, cilia assembly and disassembly regulators, and summarized the new findings of the primary cilium in tumor microenvironments and different cancers, highlighting novel possibilities for therapeutic target in cancer.
Collapse
Affiliation(s)
- Bo Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zheyong Liang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Guen VJ, Prigent C. Targeting Primary Ciliogenesis with Small-Molecule Inhibitors. Cell Chem Biol 2020; 27:1224-1228. [PMID: 32795416 DOI: 10.1016/j.chembiol.2020.07.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/12/2020] [Accepted: 07/22/2020] [Indexed: 11/18/2022]
Abstract
The primary cilium is generally a non-motile solitary organelle that protrudes from a basal body at the cell surface in various cell types in multicellular organisms. This microtubule-based structure acts as a cell signaling platform to control key cellular processes, including cell proliferation and differentiation in development and in adult tissues. Elongated and/or dysfunctional primary cilia cause developmental disorders termed ciliopathies and cancers. The genetic inhibition of ciliogenesis inducers can block the progression of these diseases in model organisms. Thus, pharmacological inhibition of primary ciliogenesis has emerged as a potential strategy to treat these pathological conditions. Pharmacological inhibitors that affect cilium assembly, and have an impact on other cellular processes, have been identified. Here, we review some of these tools and discuss their value and limitations in the study of primary cilium biology, as well as for the treatment of some ciliopathies and cancers.
Collapse
Affiliation(s)
- Vincent J Guen
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes)- UMR 6290, 35000 Rennes, France.
| | - Claude Prigent
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes)- UMR 6290, 35000 Rennes, France
| |
Collapse
|
13
|
Liu P, Dodson M, Fang D, Chapman E, Zhang DD. NRF2 negatively regulates primary ciliogenesis and hedgehog signaling. PLoS Biol 2020; 18:e3000620. [PMID: 32053600 PMCID: PMC7043785 DOI: 10.1371/journal.pbio.3000620] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 02/26/2020] [Accepted: 01/22/2020] [Indexed: 01/24/2023] Open
Abstract
Primary cilia are lost during cancer development, but the mechanism regulating cilia degeneration is not determined. While transcription factor nuclear factor-erythroid 2-like 2 (NRF2) protects cells from oxidative, proteotoxic, and metabolic stress in normal cells, hyperactivation of NRF2 is oncogenic, although the detailed molecular mechanisms by which uncontrolled NRF2 activation promotes cancer progression remain unclear. Here, we report that NRF2 suppresses hedgehog (Hh) signaling through Patched 1 (PTCH1) and primary ciliogenesis via p62/sequestosome 1 (SQSTM1). PTCH1, a negative regulator of Hh signaling, is an NRF2 target gene, and as such, hyperactivation of NRF2 impairs Hh signaling. NRF2 also suppresses primary cilia formation through p62-dependent inclusion body formation and blockage of Bardet–Biedl syndrome 4 (BBS4) entrance into cilia. Simultaneous ablation of PTCH1 and p62 completely abolishes NRF2-mediated inhibition of both primary ciliogenesis and Hh signaling. Our findings reveal a previously unidentified role of NRF2 in controlling a cellular organelle, the primary cilium, and its associated Hh signaling pathway and also uncover a mechanism by which NRF2 hyperactivation promotes tumor progression via primary cilia degeneration and aberrant Hh signaling. A better understanding of the crosstalk between NRF2 and primary cilia/Hh signaling could not only open new avenues for cancer therapeutic discovery but could also have significant implications regarding pathologies other than cancer, including developmental disorders, in which improper primary ciliogenesis and Hh signaling play a major role. This study reveals a novel role for the transcription factor NRF2 in controlling the primary cilium and its associated Hedgehog signaling pathway and also uncovers a mechanism by which NRF2 hyperactivation promotes tumor progression.
Collapse
Affiliation(s)
- Pengfei Liu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, United States of America
| | - Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, United States of America
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, United States of America
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, United States of America.,The University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
14
|
Wijaya J, Vo BT, Liu J, Xu B, Wu G, Wang Y, Peng J, Zhang J, Janke LJ, Orr BA, Yu J, Roussel MF, Schuetz JD. An ABC Transporter Drives Medulloblastoma Pathogenesis by Regulating Sonic Hedgehog Signaling. Cancer Res 2020; 80:1524-1537. [PMID: 31948942 DOI: 10.1158/0008-5472.can-19-2054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/05/2019] [Accepted: 01/07/2020] [Indexed: 01/22/2023]
Abstract
Mutations in Sonic hedgehog (SHH) signaling promote aberrant proliferation and tumor growth. SHH-medulloblastoma (MB) is among the most frequent brain tumors in children less than 3 years of age. Although key components of the SHH pathway are well-known, we hypothesized that new disease-modifying targets of SHH-MB might be identified from large-scale bioinformatics and systems biology analyses. Using a data-driven systems biology approach, we built a MB-specific interactome. The ATP-binding cassette transporter ABCC4 was identified as a modulator of SHH-MB. Accordingly, increased ABCC4 expression correlated with poor overall survival in patients with SHH-MB. Knockdown of ABCC4 expression markedly blunted the constitutive activation of the SHH pathway secondary to Ptch1 or Sufu insufficiency. In human tumor cell lines, ABCC4 knockdown and inhibition reduced full-length GLI3 levels. In a clinically relevant murine SHH-MB model, targeted ablation of Abcc4 in primary tumors significantly reduced tumor burden and extended the lifespan of tumor-bearing mice. These studies reveal ABCC4 as a potent SHH pathway regulator and a new candidate to target with the potential to improve SHH-MB therapy. SIGNIFICANCE: These findings identify ABCC4 transporter as a new target in SHH-MB, prompting the development of inhibitors or the repurporsing of existing drugs to target ABCC4.
Collapse
Affiliation(s)
- Juwina Wijaya
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - BaoHan T Vo
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jingjing Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Gang Wu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yao Wang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Junmin Peng
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee.,Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, California
| | - Laura J Janke
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Martine F Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John D Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
15
|
Zheng L, Rui C, Zhang H, Chen J, Jia X, Xiao Y. Sonic hedgehog signaling in epithelial tissue development. Regen Med Res 2019; 7:3. [PMID: 31898580 PMCID: PMC6941452 DOI: 10.1051/rmr/190004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022] Open
Abstract
The Sonic hedgehog (SHH) signaling pathway is essential for embryonic development and tissue regeneration. The dysfunction of SHH pathway is involved in a variety of diseases, including cancer, birth defects, and other diseases. Here we reviewed recent studies on main molecules involved in the SHH signaling pathway, specifically focused on their function in epithelial tissue and appendages development, including epidermis, touch dome, hair, sebaceous gland, mammary gland, tooth, nail, gastric epithelium, and intestinal epithelium. The advance in understanding the SHH signaling pathway will give us more clues to the mechanisms of tissue repair and regeneration, as well as the development of new treatment for diseases related to dysregulation of SHH signaling pathway.
Collapse
Affiliation(s)
- Lu Zheng
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Chen Rui
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Hao Zhang
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Jing Chen
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Xiuzhi Jia
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| | - Ying Xiao
-
Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University Hangzhou PR China
| |
Collapse
|
16
|
Galperin I, Dempwolff L, Diederich WE, Lauth M. Inhibiting Hedgehog: An Update on Pharmacological Compounds and Targeting Strategies. J Med Chem 2019; 62:8392-8411. [DOI: 10.1021/acs.jmedchem.9b00188] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ilya Galperin
- Center for Tumor and Immune Biology (ZTI), Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Lukas Dempwolff
- School of Pharmacy, Center for Tumor and Immune Biology (ZTI), Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Wibke E. Diederich
- School of Pharmacy, Center for Tumor and Immune Biology (ZTI), Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
- Core Facility Medicinal Chemistry, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Matthias Lauth
- Center for Tumor and Immune Biology (ZTI), Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| |
Collapse
|
17
|
Triptolide suppresses pancreatic cancer cell proliferation by inhibiting hedgehog signaling pathway activity. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1409-1412. [DOI: 10.1007/s11427-018-9477-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/03/2019] [Indexed: 10/27/2022]
|
18
|
Pietrobono S, Stecca B. Targeting the Oncoprotein Smoothened by Small Molecules: Focus on Novel Acylguanidine Derivatives as Potent Smoothened Inhibitors. Cells 2018; 7:cells7120272. [PMID: 30558232 PMCID: PMC6316656 DOI: 10.3390/cells7120272] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022] Open
Abstract
Hedgehog-GLI (HH) signaling was originally identified as a critical morphogenetic pathway in embryonic development. Since its discovery, a multitude of studies have reported that HH signaling also plays key roles in a variety of cancer types and in maintaining tumor-initiating cells. Smoothened (SMO) is the main transducer of HH signaling, and in the last few years, it has emerged as a promising therapeutic target for anticancer therapy. Although vismodegib and sonidegib have demonstrated effectiveness for the treatment of basal cell carcinoma (BCC), their clinical use has been hampered by severe side effects, low selectivity against cancer stem cells, and the onset of mutation-driven drug resistance. Moreover, SMO antagonists are not effective in cancers where HH activation is due to mutations of pathway components downstream of SMO, or in the case of noncanonical, SMO-independent activation of the GLI transcription factors, the final mediators of HH signaling. Here, we review the current and rapidly expanding field of SMO small-molecule inhibitors in experimental and clinical settings, focusing on a class of acylguanidine derivatives. We also discuss various aspects of SMO, including mechanisms of resistance to SMO antagonists.
Collapse
Affiliation(s)
- Silvia Pietrobono
- Tumor Cell Biology Unit⁻Core Research Laboratory, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy.
| | - Barbara Stecca
- Tumor Cell Biology Unit⁻Core Research Laboratory, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy.
| |
Collapse
|
19
|
Khatra H, Khan PP, Pattanayak S, Bhadra J, Rather B, Chakrabarti S, Saha T, Sinha S. Hedgehog Antagonist Pyrimidine-Indole Hybrid Molecule Inhibits Ciliogenesis through Microtubule Destabilisation. Chembiochem 2018; 19:723-735. [PMID: 29363254 DOI: 10.1002/cbic.201700631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Indexed: 12/12/2022]
Abstract
One of the crucial regulators of embryonic patterning and tissue development is the Hedgehog-glioma (Hh-Gli) signalling pathway; its uncontrolled activation has been implicated in different types of cancer in adult tissues. Primary cilium is one of the important factors required for the activation of Hh signalling, as it brings the critical components together for key protein-protein interactions required for Hh pathway regulation. Most of the synthetic and natural small molecule modulators of the pathway primarily antagonise Smoothened (Smo) or other effectors like Hh ligand or Gli. Here, we report a previously described Hh antagonist, with a pyrimidine-indole hybrid (PIH) core structure, as an inhibitor of ciliogenesis. The compound is unique in its mode of action, as it shows perturbation of microtubule dynamics in both cell-based assays and in vivo systems (zebrafish embryos). Further studies revealed that the probable targets are α-tubulin and its acetylated form, found in the cytoplasm and primary cilia. PIH also showed axonal defasiculation in developing zebrafish embryos. We thus propose that PIH antagonises Hh signalling by repressing cilia biogenesis and disassembling α-tubulin from its stabilised form.
Collapse
Affiliation(s)
- Harleen Khatra
- Department of Organic Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700032, India
| | - Pragya Paramita Khan
- Department of Organic Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700032, India
| | - Sankha Pattanayak
- Department of Organic Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700032, India
| | - Jhuma Bhadra
- Department of Organic Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700032, India
| | - Bilal Rather
- Structural Biology and Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, 700 032, India
| | - Saikat Chakrabarti
- Structural Biology and Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, 700 032, India
| | - Taniya Saha
- Division of Molecular Medicine, Bose Institute, Kolkata, 700 009, India
| | - Surajit Sinha
- Department of Organic Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700032, India
| |
Collapse
|
20
|
Gencer S, Oleinik N, Kim J, Panneer Selvam S, De Palma R, Dany M, Nganga R, Thomas RJ, Senkal CE, Howe PH, Ogretmen B. TGF-β receptor I/II trafficking and signaling at primary cilia are inhibited by ceramide to attenuate cell migration and tumor metastasis. Sci Signal 2017; 10:eaam7464. [PMID: 29066540 PMCID: PMC5818989 DOI: 10.1126/scisignal.aam7464] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Signaling by the transforming growth factor-β (TGF-β) receptors I and II (TβRI/II) and the primary cilia-localized sonic hedgehog (Shh) pathway promote cell migration and, consequently, tumor metastasis. In contrast, the sphingolipid ceramide inhibits cell proliferation and tumor metastasis. We investigated whether ceramide metabolism inhibited TβRI/II trafficking to primary cilia to attenuate cross-talk between TβRI/II and the Shh pathway. We found that ceramide synthase 4 (CerS4)-generated ceramide stabilized the association between TβRI and the inhibitory factor Smad7, which limited the trafficking of TβRI/II to primary cilia. Expression of a mutant TβRI that signals but does not interact with Smad7 prevented the CerS4-mediated inhibition of migration in various cancer cells. Genetic deletion or knockdown of CerS4 prevented the formation of the Smad7-TβRI inhibitory complex and increased the association between TβRI and the transporter Arl6 through a previously unknown cilia-targeting signal (Ala31Thr32Ala33Leu34Gln35) in TβRI. Mutating the cilia-targeting signal abolished the trafficking of TβRI to the primary cilia. Localization of TβRI to primary cilia activated a key mediator of Shh signaling, Smoothened (Smo), which stimulated cellular migration and invasion. TβRI-Smo cross-talk at the cilia in CerS4-deficient 4T1 mammary cancer cells induced liver metastasis from orthotopic allografts in both wild-type and CerS4-deficient mice, which was prevented by overexpression of Smad7 or knockdown of intraflagellar transport protein 88 (IFT88). Overall, these data reveal a ceramide-dependent mechanism that suppresses cell migration and invasion by restricting TβRI/II-Shh signaling selectively at the plasma membrane of the primary cilium.
Collapse
Affiliation(s)
- Salih Gencer
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Natalia Oleinik
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Jisun Kim
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Shanmugam Panneer Selvam
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Ryan De Palma
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Mohammed Dany
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Rose Nganga
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Raquela J Thomas
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Can E Senkal
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 125 Ashley Avenue, Charleston, SC 29425, USA.
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| |
Collapse
|
21
|
Wu F, Zhang Y, Sun B, McMahon AP, Wang Y. Hedgehog Signaling: From Basic Biology to Cancer Therapy. Cell Chem Biol 2017; 24:252-280. [PMID: 28286127 DOI: 10.1016/j.chembiol.2017.02.010] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/29/2016] [Accepted: 02/10/2017] [Indexed: 02/07/2023]
Abstract
The Hedgehog (HH) signaling pathway was discovered originally as a key pathway in embryonic patterning and development. Since its discovery, it has become increasingly clear that the HH pathway also plays important roles in a multitude of cancers. Therefore, HH signaling has emerged as a therapeutic target of interest for cancer therapy. In this review, we provide a brief overview of HH signaling and the key molecular players involved and offer an up-to-date summary of our current knowledge of endogenous and exogenous small molecules that modulate HH signaling. We discuss experiences and lessons learned from the decades-long efforts toward the development of cancer therapies targeting the HH pathway. Challenges to develop next-generation cancer therapies are highlighted.
Collapse
Affiliation(s)
- Fujia Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
22
|
Dyson JM, Conduit SE, Feeney SJ, Hakim S, DiTommaso T, Fulcher AJ, Sriratana A, Ramm G, Horan KA, Gurung R, Wicking C, Smyth I, Mitchell CA. INPP5E regulates phosphoinositide-dependent cilia transition zone function. J Cell Biol 2016; 216:247-263. [PMID: 27998989 PMCID: PMC5223597 DOI: 10.1083/jcb.201511055] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 09/19/2016] [Accepted: 12/02/2016] [Indexed: 02/07/2023] Open
Abstract
Dyson et al. demonstrate that the inositol polyphosphate 5-phosphatase INPP5E is essential for Hedgehog-dependent embryonic development. By regulating PI(4,5)P2 and PI(3,4,5)P3 signals at cilia, INPP5E contributes to cilia transition zone function and thereby Smoothened accumulation at cilia. Human ciliopathies, including Joubert syndrome (JBTS), arise from cilia dysfunction. The inositol polyphosphate 5-phosphatase INPP5E localizes to cilia and is mutated in JBTS. Murine Inpp5e ablation is embryonically lethal and recapitulates JBTS, including neural tube defects and polydactyly; however, the underlying defects in cilia signaling and the function of INPP5E at cilia are still emerging. We report Inpp5e−/− embryos exhibit aberrant Hedgehog-dependent patterning with reduced Hedgehog signaling. Using mouse genetics, we show increasing Hedgehog signaling via Smoothened M2 expression rescues some Inpp5e−/− ciliopathy phenotypes and “normalizes” Hedgehog signaling. INPP5E’s phosphoinositide substrates PI(4,5)P2 and PI(3,4,5)P3 accumulated at the transition zone (TZ) in Hedgehog-stimulated Inpp5e−/− cells, which was associated with reduced recruitment of TZ scaffolding proteins and reduced Smoothened levels at cilia. Expression of wild-type, but not 5-phosphatase-dead, INPP5E restored TZ molecular organization and Smoothened accumulation at cilia. Therefore, we identify INPP5E as an essential point of convergence between Hedgehog and phosphoinositide signaling at cilia that maintains TZ function and Hedgehog-dependent embryonic development.
Collapse
Affiliation(s)
- Jennifer M Dyson
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Sarah E Conduit
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Sandra J Feeney
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Sandra Hakim
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Tia DiTommaso
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Absorn Sriratana
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Georg Ramm
- Monash Micro Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Kristy A Horan
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Rajendra Gurung
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Carol Wicking
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ian Smyth
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia.,Development and Stem Cells Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Christina A Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
23
|
Lee H, Ko HW. Ciliary smoothened-mediated noncanonical hedgehog signaling promotes tubulin acetylation. Biochem Biophys Res Commun 2016; 480:574-579. [PMID: 27793670 DOI: 10.1016/j.bbrc.2016.10.093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/24/2016] [Indexed: 12/12/2022]
Abstract
Hedgehog (Hh) signaling plays key roles in animal development and tissue homeostasis. Binding of the secreted ligand to its Ptch1 receptor triggers Hh signaling through distinct canonical or noncanonical signaling pathways. Canonical Hh signaling leads to the activation of Gli transcription factors to induce Hh target-gene expression. In contrast, noncanonical Hh signaling regulates cytoskeleton rearrangement and apoptosis. Recently, it has been shown that primary cilia are important for canonical Hh signaling, but the ciliary role for signaling through the noncanonical pathway remains unresolved. Here, we examine the role of primary cilia in noncanonical Hh signaling in cultured mammalian cells. We found that Hh pathway activation in mouse embryonic fibroblast cells (MEFs) increases microtubule acetylation via smoothened (Smo), and suppression of Hh signaling by a Smo antagonist abrogates the microtubule acetylation. Using genetically engineered MEFs, we revealed that the increase in microtubule acetylation by Hh is dependent on Smo, but not on Sufu or Gli. In Kif3a-/- MEFs, which cannot form primary cilia, we observed that primary cilia were required for transducing noncanonical Hh signaling. Furthermore, we revealed that an increase in intracellular calcium is important for Hh-dependent tubulin acetylation at the downstream of Smo. Collectively, these findings suggest that Smo and primary cilia-dependent noncanonical Hh signaling leads to post-translational regulation of microtubules and may be important for modulating cell behaviors.
Collapse
Affiliation(s)
- Hankyu Lee
- College of Pharmacy, Dongguk University-Seoul, 32 Donggukro, Ilsandonggu, Goyangsi, Gyeonggido, 10326, South Korea
| | - Hyuk Wan Ko
- College of Pharmacy, Dongguk University-Seoul, 32 Donggukro, Ilsandonggu, Goyangsi, Gyeonggido, 10326, South Korea.
| |
Collapse
|
24
|
Maschinot CA, Pace JR, Hadden MK. Synthetic Small Molecule Inhibitors of Hh Signaling As Anti-Cancer Chemotherapeutics. Curr Med Chem 2016; 22:4033-57. [PMID: 26310919 DOI: 10.2174/0929867322666150827093904] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 12/11/2022]
Abstract
The hedgehog (Hh) pathway is a developmental signaling pathway that is essential to the proper embryonic development of many vertebrate systems. Dysregulation of Hh signaling has been implicated as a causative factor in the development and progression of several forms of human cancer. As such, the development of small molecule inhibitors of Hh signaling as potential anti-cancer chemotherapeutics has been a major area of research interest in both academics and industry over the past ten years. Through these efforts, synthetic small molecules that target multiple components of the Hh pathway have been identified and advanced to preclinical or clinical development. The goal of this review is to provide an update on the current status of several synthetic small molecule Hh pathway inhibitors and explore the potential of several recently disclosed inhibitory scaffolds.
Collapse
Affiliation(s)
| | | | - M K Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092, USA.
| |
Collapse
|
25
|
Jung B, Messias AC, Schorpp K, Geerlof A, Schneider G, Saur D, Hadian K, Sattler M, Wanker EE, Hasenöder S, Lickert H. Novel small molecules targeting ciliary transport of Smoothened and oncogenic Hedgehog pathway activation. Sci Rep 2016; 6:22540. [PMID: 26931153 PMCID: PMC4773810 DOI: 10.1038/srep22540] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/15/2016] [Indexed: 01/04/2023] Open
Abstract
Trafficking of the G protein-coupled receptor (GPCR) Smoothened (Smo) to the primary cilium (PC) is a potential target to inhibit oncogenic Hh pathway activation in a large number of tumors. One drawback is the appearance of Smo mutations that resist drug treatment, which is a common reason for cancer treatment failure. Here, we undertook a high content screen with compounds in preclinical or clinical development and identified ten small molecules that prevent constitutive active mutant SmoM2 transport into PC for subsequent Hh pathway activation. Eight of the ten small molecules act through direct interference with the G protein-coupled receptor associated sorting protein 2 (Gprasp2)-SmoM2 ciliary targeting complex, whereas one antagonist of ionotropic receptors prevents intracellular trafficking of Smo to the PC. Together, these findings identify several compounds with the potential to treat drug-resistant SmoM2-driven cancer forms, but also reveal off-target effects of established drugs in the clinics.
Collapse
Affiliation(s)
- Bomi Jung
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany
| | - Ana C Messias
- Institute of Structural Biology, Helmholtz Zentrum München, Germany.,Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department Chemistry, Technische Universität München, 85747 Garching, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Helmholtz Zentrum München, Germany
| | - Arie Geerlof
- Institute of Structural Biology, Helmholtz Zentrum München, Germany
| | - Günter Schneider
- Department of Internal Medicine II, Klinikum rechts der Isar, München, Germany.,Technische Universität München, München, Germany
| | - Dieter Saur
- Department of Internal Medicine II, Klinikum rechts der Isar, München, Germany.,Technische Universität München, München, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Helmholtz Zentrum München, Germany
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Germany.,Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department Chemistry, Technische Universität München, 85747 Garching, Germany
| | - Erich E Wanker
- Neuroproteomics, Max Delbrueck Center for Molecular Medicine, 13125 Berlin, Germany
| | - Stefan Hasenöder
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum München, Germany.,Technische Universität München, München, Germany.,German Center for Diabetes Research (DZD), Germany
| |
Collapse
|
26
|
Jung B, Padula D, Burtscher I, Landerer C, Lutter D, Theis F, Messias AC, Geerlof A, Sattler M, Kremmer E, Boldt K, Ueffing M, Lickert H. Pitchfork and Gprasp2 Target Smoothened to the Primary Cilium for Hedgehog Pathway Activation. PLoS One 2016; 11:e0149477. [PMID: 26901434 PMCID: PMC4763541 DOI: 10.1371/journal.pone.0149477] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/31/2016] [Indexed: 01/14/2023] Open
Abstract
The seven-transmembrane receptor Smoothened (Smo) activates all Hedgehog (Hh) signaling by translocation into the primary cilia (PC), but how this is regulated is not well understood. Here we show that Pitchfork (Pifo) and the G protein-coupled receptor associated sorting protein 2 (Gprasp2) are essential components of an Hh induced ciliary targeting complex able to regulate Smo translocation to the PC. Depletion of Pifo or Gprasp2 leads to failure of Smo translocation to the PC and lack of Hh target gene activation. Together, our results identify a novel protein complex that is regulated by Hh signaling and required for Smo ciliary trafficking and Hh pathway activation.
Collapse
Affiliation(s)
- Bomi Jung
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, München-Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Daniela Padula
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, München-Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, München-Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Cedric Landerer
- Institute of Computational Biology, Helmholtz Zentrum München, München-Neuherberg, Germany
- Department of Ecology & Evolutionary Biology, University of Tennessee, Knoxville TN 37996, United States of America
| | - Dominik Lutter
- Institute of Computational Biology, Helmholtz Zentrum München, München-Neuherberg, Germany
- Institute of Diabetes and Adipositas, Helmholtz Zentrum München, München-Neuherberg, Germany
| | - Fabian Theis
- Institute of Computational Biology, Helmholtz Zentrum München, München-Neuherberg, Germany
| | - Ana C. Messias
- Institute of Structural Biology, Helmholtz Zentrum München, München-Neuherberg, Germany
- Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, 85747 Garching, Germany
| | - Arie Geerlof
- Institute of Structural Biology, Helmholtz Zentrum München, München-Neuherberg, Germany
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, München-Neuherberg, Germany
- Center for Integrated Protein Science Munich at Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, 85747 Garching, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, München-Neuherberg, Germany
| | - Karsten Boldt
- Department of Protein Science, Helmholtz Zentrum München, München-Neuherberg, Germany
- Centre of Ophthalmology, Institute for Ophthalmology Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Marius Ueffing
- Department of Protein Science, Helmholtz Zentrum München, München-Neuherberg, Germany
- Centre of Ophthalmology, Institute for Ophthalmology Research, University of Tuebingen, 72076 Tuebingen, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, München-Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
- * E-mail:
| |
Collapse
|
27
|
Owen TS, Ngoje G, Lageman TJ, Bordeau BM, Belfort M, Callahan BP. Förster resonance energy transfer-based cholesterolysis assay identifies a novel hedgehog inhibitor. Anal Biochem 2015; 488:1-5. [PMID: 26095399 DOI: 10.1016/j.ab.2015.06.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/12/2015] [Accepted: 06/12/2015] [Indexed: 01/20/2023]
Abstract
Hedgehog (Hh) proteins function in cell/cell signaling processes linked to human embryo development and the progression of several types of cancer. Here, we describe an optical assay of hedgehog cholesterolysis, a unique autoprocessing event critical for Hh function. The assay uses a recombinant Förster resonance energy transfer (FRET)-active Hh precursor whose cholesterolysis can be monitored continuously in multi-well plates (dynamic range=4, Z'=0.7), offering advantages in throughput over conventional sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) assays. Application of the optical assay in a pilot small molecule screen produced a novel cholesterolysis inhibitor (apparent IC50=5×10(-6)M) that appears to inactivate hedgehog covalently by a substitution nucleophilic aromatic (SNAr) mechanism.
Collapse
Affiliation(s)
- Timothy S Owen
- Department of Chemistry, Binghamton University, Binghamton, NY 13902, USA
| | - George Ngoje
- Department of Chemistry, Binghamton University, Binghamton, NY 13902, USA
| | - Travis J Lageman
- Department of Chemistry, Binghamton University, Binghamton, NY 13902, USA
| | - Brandon M Bordeau
- Department of Chemistry, Binghamton University, Binghamton, NY 13902, USA
| | - Marlene Belfort
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Brian P Callahan
- Department of Chemistry, Binghamton University, Binghamton, NY 13902, USA.
| |
Collapse
|
28
|
Finetti F, Patrussi L, Galgano D, Cassioli C, Perinetti G, Pazour GJ, Baldari CT. The small GTPase Rab8 interacts with VAMP-3 to regulate the delivery of recycling T-cell receptors to the immune synapse. J Cell Sci 2015; 128:2541-52. [PMID: 26034069 DOI: 10.1242/jcs.171652] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/22/2015] [Indexed: 12/31/2022] Open
Abstract
IFT20, a component of the intraflagellar transport (IFT) system that controls ciliogenesis, regulates immune synapse assembly in the non-ciliated T-cell by promoting T-cell receptor (TCR) recycling. Here, we have addressed the role of Rab8 (for which there are two isoforms Rab8a and Rab8b), a small GTPase implicated in ciliogenesis, in TCR traffic to the immune synapse. We show that Rab8, which colocalizes with IFT20 in Rab11(+) endosomes, is required for TCR recycling. Interestingly, as opposed to in IFT20-deficient T-cells, TCR(+) endosomes polarized normally beneath the immune synapse membrane in the presence of dominant-negative Rab8, but were unable to undergo the final docking or fusion step. This could be accounted for by the inability of the vesicular (v)-SNARE VAMP-3 to cluster at the immune synapse in the absence of functional Rab8, which is responsible for its recruitment. Of note, and similar to in T-cells, VAMP-3 interacts with Rab8 at the base of the cilium in NIH-3T3 cells, where it regulates ciliary growth and targeting of the protein smoothened. The results identify Rab8 as a new player in vesicular traffic to the immune synapse and provide insight into the pathways co-opted by different cell types for immune synapse assembly and ciliogenesis.
Collapse
Affiliation(s)
- Francesca Finetti
- Department of Life Sciences, University of Siena, Siena 53100, Italy
| | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena 53100, Italy
| | - Donatella Galgano
- Department of Life Sciences, University of Siena, Siena 53100, Italy
| | - Chiara Cassioli
- Department of Life Sciences, University of Siena, Siena 53100, Italy
| | - Giuseppe Perinetti
- Department of Medical, Surgical and Health Sciences, School of Dentistry, University of Trieste, Trieste 34129, Italy
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, Siena 53100, Italy
| |
Collapse
|
29
|
Onnis A, Finetti F, Patrussi L, Gottardo M, Cassioli C, Spanò S, Baldari CT. The small GTPase Rab29 is a common regulator of immune synapse assembly and ciliogenesis. Cell Death Differ 2015; 22:1687-99. [PMID: 26021297 DOI: 10.1038/cdd.2015.17] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 01/21/2015] [Accepted: 02/02/2015] [Indexed: 01/09/2023] Open
Abstract
Accumulating evidence underscores the T-cell immune synapse (IS) as a site of intense vesicular trafficking, on which productive signaling and cell activation crucially depend. Although the T-cell antigen receptor (TCR) is known to exploit recycling to accumulate to the IS, the specific pathway that controls this process remains to be elucidated. Here we demonstrate that the small GTPase Rab29 is centrally implicated in TCR trafficking and IS assembly. Rab29 colocalized and interacted with Rab8, Rab11 and IFT20, a component of the intraflagellar transport system that regulates ciliogenesis and participates in TCR recycling in the non-ciliated T cell, as assessed by co-immunoprecipitation and immunofluorescence analysis. Rab29 depletion resulted in the inability of TCRs to undergo recycling to the IS, thereby compromizing IS assembly. Under these conditions, recycling TCRs accumulated in Rab11(+) endosomes that failed to polarize to the IS due to defective Rab29-dependent recruitment of the dynein microtubule motor. Remarkably, Rab29 participates in a similar pathway in ciliated cells to promote primary cilium growth and ciliary localization of Smoothened. These results provide a function for Rab29 as a regulator of receptor recycling and identify this GTPase as a shared participant in IS and primary cilium assembly.
Collapse
Affiliation(s)
- A Onnis
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - F Finetti
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - L Patrussi
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - M Gottardo
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - C Cassioli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - S Spanò
- School of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - C T Baldari
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| |
Collapse
|
30
|
Larsen AR, Bai RY, Chung JH, Borodovsky A, Rudin CM, Riggins GJ, Bunz F. Repurposing the antihelmintic mebendazole as a hedgehog inhibitor. Mol Cancer Ther 2014; 14:3-13. [PMID: 25376612 DOI: 10.1158/1535-7163.mct-14-0755-t] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The hedgehog (Hh) signaling pathway is activated in many types of cancer and therefore presents an attractive target for new anticancer agents. Here, we show that mebendazole, a benzamidazole with a long history of safe use against nematode infestations and hydatid disease, potently inhibited Hh signaling and slowed the growth of Hh-driven human medulloblastoma cells at clinically attainable concentrations. As an antiparasitic, mebendazole avidly binds nematode tubulin and causes inhibition of intestinal microtubule synthesis. In human cells, mebendazole suppressed the formation of the primary cilium, a microtubule-based organelle that functions as a signaling hub for Hh pathway activation. The inhibition of Hh signaling by mebendazole was unaffected by mutants in the gene that encodes human Smoothened (SMO), which are selectively propagated in cell clones that survive treatment with the Hh inhibitor vismodegib. Combination of vismodegib and mebendazole resulted in additive Hh signaling inhibition. Because mebendazole can be safely administered to adults and children at high doses over extended time periods, we propose that mebendazole could be rapidly repurposed and clinically tested as a prospective therapeutic agent for many tumors that are dependent on Hh signaling.
Collapse
Affiliation(s)
- Andrew R Larsen
- Department of Radiation Oncology and Molecular Radiation Sciences, The Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Ren-Yuan Bai
- Department of Neurosurgery, The Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Jon H Chung
- Department of Radiation Oncology and Molecular Radiation Sciences, The Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Alexandra Borodovsky
- Department of Neurosurgery, The Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Charles M Rudin
- Memorial Hospital Research Laboratories, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gregory J Riggins
- Department of Neurosurgery, The Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland.
| | - Fred Bunz
- Department of Radiation Oncology and Molecular Radiation Sciences, The Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland.
| |
Collapse
|
31
|
Hedgehog pathway inhibitor-4 suppresses malignant properties of chondrosarcoma cells by disturbing tumor ciliogenesis. Oncol Rep 2014; 32:1622-30. [PMID: 25110171 DOI: 10.3892/or.2014.3372] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/08/2014] [Indexed: 11/05/2022] Open
Abstract
Chondrosarcoma is a type of malignant bone tumor secreting cartilage-like matrix. In clinical treatment, there is no frequently used drug treatment option except for surgical resection. Hedgehog (HH) pathway is a classical signaling pathway that regulates normal cartilage cell development. In order to detect the role that HH pathway plays in chondrosarcoma, we used immunohistochemistry and found this tumor clearly expressed HH pathway-related proteins. Treatment with HH pathway inhibitor-4 (HPI-4) could significantly decrease human chondrosarcoma cell proliferation, invasion and migration ability. Furthermore, HPI-4 could distinctly disturb HH pathway-mediated ciliogenesis and suppress primary cilia-related protein intraflagellar transport protein IFT88 expression. HH downstream effect molecular GLI2 was restrained to block parathyroid hormone-related protein and affect MAPK/ERK-regulated matrix metalloproteinases (MMP2 and MMP9). These results indicated that activated HH pathway existed in chondrosarcoma and HPI-4 could be a new therapeutic option specific to chondrosarcoma expressing elevated levels of HH pathway.
Collapse
|
32
|
Structural insights into the role of the Smoothened cysteine-rich domain in Hedgehog signalling. Nat Commun 2014; 4:2965. [PMID: 24351982 PMCID: PMC3890372 DOI: 10.1038/ncomms3965] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 11/20/2013] [Indexed: 01/09/2023] Open
Abstract
Smoothened (Smo) is a member of the Frizzled (FzD) class of G-protein-coupled receptors (GPCRs), and functions as the key transducer in the Hedgehog (Hh) signalling pathway. Smo has an extracellular cysteine-rich domain (CRD), indispensable for its function and downstream Hh signalling. Despite its essential role, the functional contribution of the CRD to Smo signalling has not been clearly elucidated. However, given that the FzD CRD binds to the endogenous Wnt ligand, it has been proposed that the Smo CRD may bind its own endogenous ligand. Here we present the NMR solution structure of the Drosophila Smo CRD, and describe interactions between the glucocorticoid budesonide (Bud) and the Smo CRDs from both Drosophila and human. Our results highlight a function of the Smo CRD, demonstrating its role in binding to small-molecule modulators.
Collapse
|
33
|
Banerjee U, Hadden MK. Recent advances in the design of Hedgehog pathway inhibitors for the treatment of malignancies. Expert Opin Drug Discov 2014; 9:751-71. [PMID: 24850423 DOI: 10.1517/17460441.2014.920817] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION The Hedgehog (Hh) signaling pathway is known to be dysregulated in several forms of cancer. Hence, specifically targeting this signaling cascade is a valid and promising strategy for successful therapeutic intervention. Several components within the Hh pathway have been proven to be druggable; however, challenges in the discovery and development process for small molecules targeting this pathway have been identified. AREAS COVERED This review details both the current state and future potential of Hh pathway inhibitors as anticancer chemotherapeutics that target a variety of human malignancies. EXPERT OPINION The initial development of Hh pathway inhibitors focused on small-molecule antagonists of Smoothened, a transmembrane protein that is a key regulator of pathway signaling. More recently, efforts to identify and develop inhibitors of pathway signaling that function through alternate mechanisms have been increasing. However, none of these have advanced into clinical trials. Further, early evidence suggesting the broad application of Hh pathway inhibitors as a monotherapy in a wide range of human cancers has not been validated. The potential for Hh pathway inhibitors as combination therapy has demonstrated promising preclinical results. However, more research to identify rational drug combinations to fully explore the potential of this anticancer drug class is warranted.
Collapse
Affiliation(s)
- Upasana Banerjee
- University of Connecticut, Department of Pharmaceutical Sciences , 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092 , USA +1 860 486 8446 ;
| | | |
Collapse
|
34
|
G-protein-coupled receptors, Hedgehog signaling and primary cilia. Semin Cell Dev Biol 2014; 33:63-72. [PMID: 24845016 DOI: 10.1016/j.semcdb.2014.05.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/12/2014] [Indexed: 12/21/2022]
Abstract
The Hedgehog (Hh) pathway has become an important model to study the cell biology of primary cilia, and reciprocally, the study of ciliary processes provides an opportunity to solve longstanding mysteries in the mechanism of vertebrate Hh signal transduction. The cilium is emerging as an unique compartment for G-protein-coupled receptor (GPCR) signaling in many systems. Two members of the GPCR family, Smoothened and Gpr161, play important roles in the Hh pathway. We review the current understanding of how these proteins may function to regulate Hh signaling and also highlight some of the critical unanswered questions being tackled by the field. Uncovering GPCR-regulated mechanisms important in Hh signaling may provide therapeutic strategies against the Hh pathway that plays important roles in development, regeneration and cancer.
Collapse
|
35
|
Singh S, Carpenter AE, Genovesio A. Increasing the Content of High-Content Screening: An Overview. ACTA ACUST UNITED AC 2014; 19:640-50. [PMID: 24710339 PMCID: PMC4230961 DOI: 10.1177/1087057114528537] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 12/31/2013] [Indexed: 01/17/2023]
Abstract
Target-based high-throughput screening (HTS) has recently been critiqued for its relatively poor yield compared to phenotypic screening approaches. One type of phenotypic screening, image-based high-content screening (HCS), has been seen as particularly promising. In this article, we assess whether HCS is as high content as it can be. We analyze HCS publications and find that although the number of HCS experiments published each year continues to grow steadily, the information content lags behind. We find that a majority of high-content screens published so far (60−80%) made use of only one or two image-based features measured from each sample and disregarded the distribution of those features among each cell population. We discuss several potential explanations, focusing on the hypothesis that data analysis traditions are to blame. This includes practical problems related to managing large and multidimensional HCS data sets as well as the adoption of assay quality statistics from HTS to HCS. Both may have led to the simplification or systematic rejection of assays carrying complex and valuable phenotypic information. We predict that advanced data analysis methods that enable full multiparametric data to be harvested for entire cell populations will enable HCS to finally reach its potential.
Collapse
Affiliation(s)
- Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Auguste Genovesio
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA École Normale Supérieure, 45, Rue d'Ulm, 75005 Paris
| |
Collapse
|
36
|
Peluso MO, Campbell VT, Harari JA, Tibbitts TT, Proctor JL, Whitebread N, Conley JM, White KF, Kutok JL, Read MA, McGovern K, Faia KL. Impact of the Smoothened inhibitor, IPI-926, on smoothened ciliary localization and Hedgehog pathway activity. PLoS One 2014; 9:e90534. [PMID: 24608250 PMCID: PMC3946503 DOI: 10.1371/journal.pone.0090534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/02/2014] [Indexed: 12/31/2022] Open
Abstract
A requisite step for canonical Hedgehog (Hh) pathway activation by Sonic Hedgehog (Shh) ligand is accumulation of Smoothened (Smo) to the primary cilium (PC). Activation of the Hh pathway has been implicated in a broad range of cancers, and several Smo antagonists are being assessed clinically, one of which is approved for the treatment of advanced basal cell carcinoma. Recent reports demonstrate that various Smo antagonists differentially impact Smo localization to the PC while still exerting inhibitory activity. In contrast to other synthetic small molecule Smo antagonists, the natural product cyclopamine binds to and promotes ciliary accumulation of Smo and “primes” cells for Hh pathway hyper-responsiveness after compound withdrawal. We compared the properties of IPI-926, a semi-synthetic cyclopamine analog, to cyclopamine with regard to potency, ciliary Smo accumulation, and Hh pathway activity after compound withdrawal. Like cyclopamine, IPI-926 promoted accumulation of Smo to the PC. However, in contrast to cyclopamine, IPI-926 treatment did not prime cells for hyper-responsiveness to Shh stimulation after compound withdrawal, but instead demonstrated continuous inhibition of signaling. By comparing the levels of drug-induced ciliary Smo accumulation with the degree of Hh pathway activity after compound withdrawal, we propose that a critical threshold of ciliary Smo is necessary for “priming” activity to occur. This “priming” appears achievable with cyclopamine, but not IPI-926, and is cell-line dependent. Additionally, IPI-926 activity was evaluated in a murine tumor xenograft model and a pharmacokinetic/pharmacodynamic relationship was examined to assess for in vivo evidence of Hh pathway hyper-responsiveness. Plasma concentrations of IPI-926 correlated with the degree and duration of Hh pathway suppression, and pathway activity did not exceed baseline levels out to 96 hours post dose. The overall findings suggest that IPI-926 possesses unique biophysical and pharmacological properties that result in Hh pathway inhibition in a manner that differentiates it from cyclopamine.
Collapse
Affiliation(s)
- Marisa O. Peluso
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Veronica T. Campbell
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Joseph A. Harari
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Thomas T. Tibbitts
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Jennifer L. Proctor
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Nigel Whitebread
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - James M. Conley
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Kerry F. White
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Jeffery L. Kutok
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Margaret A. Read
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Karen McGovern
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
| | - Kerrie L. Faia
- Infinity Pharmaceuticals, Inc., Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
37
|
Sung CH, Leroux MR. The roles of evolutionarily conserved functional modules in cilia-related trafficking. Nat Cell Biol 2014; 15:1387-97. [PMID: 24296415 DOI: 10.1038/ncb2888] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cilia are present across most eukaryotic phyla and have diverse sensory and motility roles in animal physiology, cell signalling and development. Their biogenesis and maintenance depend on vesicular and intraciliary (intraflagellar) trafficking pathways that share conserved structural and functional modules. The functional units of the interconnected pathways, which include proteins involved in membrane coating as well as small GTPases and their accessory factors, were first experimentally associated with canonical vesicular trafficking. These components are, however, ancient, having been co-opted by the ancestral eukaryote to establish the ciliary organelle, and their study can inform us about ciliary biology in higher organisms.
Collapse
Affiliation(s)
- Ching-Hwa Sung
- Margaret M. Dyson Vision Research Institute, Department of Ophthalmology, Weill Medical College of Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | | |
Collapse
|
38
|
Unraveling the therapeutic potential of the Hedgehog pathway in cancer. Nat Med 2013; 19:1410-22. [PMID: 24202394 DOI: 10.1038/nm.3389] [Citation(s) in RCA: 435] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 10/01/2013] [Indexed: 02/07/2023]
Abstract
Major progress has been made in recent years in the development of Hedgehog (Hh) pathway inhibitors for the treatment of patients with cancer. Promising clinical trial results have been obtained in cancers that harbor activating mutations of the Hh pathway, such as basal cell carcinoma and medulloblastoma. However, for many cancers, in which Hh ligand overexpression is thought to drive tumor growth, results have been disappointing. Here we review the preclinical data that continue to shape our understanding of the Hh pathway in tumorigenesis and the emerging clinical experience with smoothened inhibitors.
Collapse
|
39
|
Schmidts M, Vodopiutz J, Christou-Savina S, Cortés C, McInerney-Leo A, Emes R, Arts H, Tüysüz B, D’Silva J, Leo P, Giles T, Oud M, Harris J, Koopmans M, Marshall M, Elçioglu N, Kuechler A, Bockenhauer D, Moore A, Wilson L, Janecke A, Hurles M, Emmet W, Gardiner B, Streubel B, Dopita B, Zankl A, Kayserili H, Scambler P, Brown M, Beales P, Wicking C, Duncan E, Mitchison H. Mutations in the gene encoding IFT dynein complex component WDR34 cause Jeune asphyxiating thoracic dystrophy. Am J Hum Genet 2013; 93:932-44. [PMID: 24183451 PMCID: PMC3824113 DOI: 10.1016/j.ajhg.2013.10.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 09/09/2013] [Accepted: 10/03/2013] [Indexed: 11/26/2022] Open
Abstract
Bidirectional (anterograde and retrograde) motor-based intraflagellar transport (IFT) governs cargo transport and delivery processes that are essential for primary cilia growth and maintenance and for hedgehog signaling functions. The IFT dynein-2 motor complex that regulates ciliary retrograde protein transport contains a heavy chain dynein ATPase/motor subunit, DYNC2H1, along with other less well functionally defined subunits. Deficiency of IFT proteins, including DYNC2H1, underlies a spectrum of skeletal ciliopathies. Here, by using exome sequencing and a targeted next-generation sequencing panel, we identified a total of 11 mutations in WDR34 in 9 families with the clinical diagnosis of Jeune syndrome (asphyxiating thoracic dystrophy). WDR34 encodes a WD40 repeat-containing protein orthologous to Chlamydomonas FAP133, a dynein intermediate chain associated with the retrograde intraflagellar transport motor. Three-dimensional protein modeling suggests that the identified mutations all affect residues critical for WDR34 protein-protein interactions. We find that WDR34 concentrates around the centrioles and basal bodies in mammalian cells, also showing axonemal staining. WDR34 coimmunoprecipitates with the dynein-1 light chain DYNLL1 in vitro, and mining of proteomics data suggests that WDR34 could represent a previously unrecognized link between the cytoplasmic dynein-1 and IFT dynein-2 motors. Together, these data show that WDR34 is critical for ciliary functions essential to normal development and survival, most probably as a previously unrecognized component of the mammalian dynein-IFT machinery.
Collapse
Affiliation(s)
- Miriam Schmidts
- Molecular Medicine Unit and Birth Defect Research Centre, Institute of Child Health, University College London (UCL), London WC1N 1EH, UK
| | - Julia Vodopiutz
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Sonia Christou-Savina
- Molecular Medicine Unit and Birth Defect Research Centre, Institute of Child Health, University College London (UCL), London WC1N 1EH, UK
| | - Claudio R. Cortés
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Aideen M. McInerney-Leo
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Level 7, 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | - Richard D. Emes
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, UK
- Advanced Data Analysis Centre, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, UK
| | - Heleen H. Arts
- Department of Human Genetics, Radboud University Medical Centre, Radboud University, 6500 HB Nijmegen, the Netherlands
- Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, the Netherlands
- Institute for Genetic and Metabolic Disease, Radboud University, 6500 HB Nijmegen, the Netherlands
| | - Beyhan Tüysüz
- Department of Pediatrics, Division of Pediatric Genetics, Cerrahpasa Medical Faculty, Istanbul University, 34303 Istanbul, Turkey
| | - Jason D’Silva
- Molecular Medicine Unit and Birth Defect Research Centre, Institute of Child Health, University College London (UCL), London WC1N 1EH, UK
| | - Paul J. Leo
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Level 7, 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | - Tom C. Giles
- Advanced Data Analysis Centre, University of Nottingham, Sutton Bonington Campus, Leicestershire LE12 5RD, UK
| | - Machteld M. Oud
- Department of Human Genetics, Radboud University Medical Centre, Radboud University, 6500 HB Nijmegen, the Netherlands
- Nijmegen Centre for Molecular Life Sciences, Radboud University, 6500 HB Nijmegen, the Netherlands
- Institute for Genetic and Metabolic Disease, Radboud University, 6500 HB Nijmegen, the Netherlands
| | - Jessica A. Harris
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Level 7, 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | - Marije Koopmans
- Department of Clinical Genetics, Center for Human and Clinical Genetics, Leiden University Medical Centre, 2333 AL Leiden, the Netherlands
| | - Mhairi Marshall
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Level 7, 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | - Nursel Elçioglu
- Department of Pediatrics, Marmara University Hospital, Istanbul 34716, Turkey
| | - Alma Kuechler
- Institut für Humangenetik, Universitätsklinikum Essen, Universität Duisburg-Essen, 45122 Essen, Germany
| | - Detlef Bockenhauer
- Great Ormond Street Hospital and Nephro-Urology Unit, Institute of Child Health, University College London (UCL), London WC1N 1EH, UK
| | - Anthony T. Moore
- Moorfields Eye Hospital and UCL Institute of Ophthalmology, London EC1V 2PH, UK
| | - Louise C. Wilson
- Department of Clinical Genetics, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Andreas R. Janecke
- Department of Pediatrics I, and Division of Human Genetics, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Matthew E. Hurles
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1RQ, UK
| | - Warren Emmet
- Department of Genetics, Environment and Evolution, UCL Genetics Institute (UGI), University College London, London WC1E 6BT, UK
| | - Brooke Gardiner
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Level 7, 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | - Berthold Streubel
- Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria
| | - Belinda Dopita
- Department of Genetics, The Canberra Hospital, Woden, ACT 2606, Australia
| | - Andreas Zankl
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD 4029, Australia
| | - Hülya Kayserili
- Istanbul Medical Faculty, Medical Genetics Department, Istanbul University, 34390 Istanbul, Turkey
| | - Peter J. Scambler
- Molecular Medicine Unit and Birth Defect Research Centre, Institute of Child Health, University College London (UCL), London WC1N 1EH, UK
| | - Matthew A. Brown
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Level 7, 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | - Philip L. Beales
- Molecular Medicine Unit and Birth Defect Research Centre, Institute of Child Health, University College London (UCL), London WC1N 1EH, UK
| | - Carol Wicking
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | | | - Emma L. Duncan
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, Level 7, 37 Kent Street, Woolloongabba, QLD 4102, Australia
- Department of Endocrinology, James Mayne Building, Royal Brisbane and Women’s Hospital, Butterfield Road, Herston, QLD 4029, Australia
| | - Hannah M. Mitchison
- Molecular Medicine Unit and Birth Defect Research Centre, Institute of Child Health, University College London (UCL), London WC1N 1EH, UK
| |
Collapse
|
40
|
Abstract
INTRODUCTION The hedgehog (Hh) pathway is a developmental signaling pathway that plays a key role in directing cellular growth and tissue patterning during embryonic development. Dysregulation of Hh signaling has been linked to the development of a variety of human tumors, and numerous drug development programs in both academia and industry are actively exploring inhibitors of the pathway as anti-cancer agents. AREAS COVERED This review surveys the recent patent literature (2009 - 2012) for Hh pathway inhibitors as treatments for a variety of human malignancies. EXPERT OPINION To date, all of the pathway inhibitors that have entered clinical trials and the majority of compounds identified via high-throughput screens target smoothened (Smo), a transmembrane protein that is essential for pathway signaling. While these compounds have shown initial promise in preclinical and clinical trials, several mechanisms of resistance to Smo inhibitors have been identified. Even with this knowledge, the majority of small-molecule pathway inhibitors disclosed in the recent patent literature directly target Smo. The continued identification of Hh pathway inhibitors that function either upstream or downstream is warranted not only to combat these emerging resistance mechanisms, but also to help elucidate the various cellular mechanisms that control both normal and oncogenic pathway signaling.
Collapse
Affiliation(s)
- Matthew Kyle Hadden
- Department of Pharmaceutical Sciences, University of Connecticut, 69 N Eagleville Rd, Unit 3092, Storrs, CT 06269-3092, USA
| |
Collapse
|