1
|
Huang Y, Zhao P, Zhang X, Fu H, Fu C. Uncovering the pharmacological mechanisms of Patchouli essential oil for treating ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118737. [PMID: 39182705 DOI: 10.1016/j.jep.2024.118737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pogostemonis Herba has long been used in traditional Chinese medicine to treat inflammatory disorders. Patchouli essential oil (PEO) is the primary component of Pogostemonis Herba, and it has been suggested to offer curative potential when applied to treat ulcerative colitis (UC). However, the pharmacological mechanisms of PEO for treating UC remain to be clarified. AIM OF THE STUDY To elucidate the pharmacological mechanisms of PEO for treating UC. METHODS AND RESULTS In the present study, transcriptomic and network pharmacology approaches were combined to clarify the mechanisms of PEO for treating UC. Our results reveal that rectal PEO administration in UC model mice significantly alleviated symptoms of UC. In addition, PEO effectively suppressed colonic inflammation and oxidative stress. Mechanistically, PEO can ameliorate UC mice by modulating gut microbiota, inhibiting inflammatory targets (OPTC, PTN, IFIT3, EGFR, and TLR4), and inhibiting the PI3K-AKT pathway. Next, the 11 potential bioactive components that play a role in PEO's anti-UC mechanism were identified, and the therapeutic efficacy of the pogostone (a bioactive component) in UC mice was partially validated. CONCLUSION This study highlights the mechanisms through which PEO can treat UC, providing a rigorous scientific foundation for future efforts to develop and apply PEO for treating UC.
Collapse
Affiliation(s)
- You Huang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, 611137, China
| | - Pengyu Zhao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xing Zhang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, 611137, China
| | - Hao Fu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chaomei Fu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, 611137, China.
| |
Collapse
|
2
|
Singh R, Ray A. Therapeutic potential of hedgehog signaling in advanced cancer types. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 386:49-80. [PMID: 38782501 DOI: 10.1016/bs.ircmb.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
In this chapter, we have made an attempt to elucidate the relevance of hedgehog signaling pathway in tumorigenesis. Here, we have described different types of hedgehog signaling (canonical and non-canonical) with emphasis on the different mechanisms (mutation-driven, autocrine, paracrine and reverse paracrine) it adopts during tumorigenesis. We have discussed the role of hedgehog signaling in regulating cell proliferation, invasion and epithelial-to-mesenchymal transition in both local and advanced cancer types, as reported in different studies based on preclinical and clinical models. We have specifically addressed the role of hedgehog signaling in aggressive neuroendocrine tumors as well. We have also elaborated on the studies showing therapeutic relevance of the inhibitors of hedgehog signaling in cancer. Evidence of the crosstalk of hedgehog signaling components with other signaling pathways and treatment resistance due to tumor heterogeneity have also been briefly discussed. Together, we have tried to put forward a compilation of the studies on therapeutic potential of hedgehog signaling in various cancers, specifically aggressive tumor types with a perspective into what is lacking and demands further investigation.
Collapse
Affiliation(s)
- Richa Singh
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States.
| | - Anindita Ray
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| |
Collapse
|
3
|
Genome-Wide Identification of G Protein-Coupled Receptors in Ciliated Eukaryotes. Int J Mol Sci 2023; 24:ijms24043869. [PMID: 36835283 PMCID: PMC9960496 DOI: 10.3390/ijms24043869] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/07/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of transmembrane receptors and play important roles in many physiological processes. As a representative group of protozoa, ciliates represent the highest stage of eukaryotic cell differentiation and evolution in terms of their reproductive mode, two-state karyotype, and extremely diverse cytogenesis patterns. GPCRs have been poorly reported in ciliates. In this study, we identified 492 GPCRs in 24 ciliates. Using the existing classification system for animals, GPCRs in ciliates can be assigned to four families, including families A, B, E, and F. Most (377 members) belong to family A. The number of GPCRs is extremely different in different ciliates; the Heterotrichea ciliates usually have more GPCRs than other ciliates. Parasitic or symbiotic ciliates usually have only a few GPCRs. Gene/genome duplication events seem to play important roles in the expansion of the GPCR superfamily in ciliates. GPCRs in ciliates displayed seven typical domain organizations. GPCRs in an ortholog group are common and conserved in all ciliates. The gene expression analysis of the members in this conserved ortholog group in the model ciliate, Tetrahymena thermophila, suggested that these GPCRs play important roles in the life cycle of ciliates. In summary, this study provides the first comprehensive genome-wide identification of GPCRs in ciliates, improving our understanding of the evolution and function of GPCR in ciliates.
Collapse
|
4
|
Smith AE, Sigurbjörnsdóttir ES, Steingrímsson E, Sigurbjörnsdóttir S. Hedgehog signalling in bone and osteoarthritis: the role of Smoothened and cholesterol. FEBS J 2022. [PMID: 35305060 DOI: 10.1111/febs.16440] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/25/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
Hedgehog signalling is essential for development, crucial for normal anatomical arrangement and activated during tissue damage repair. Dysregulation of hedgehog signalling is associated with cancer, developmental disorders and other diseases including osteoarthritis (OA). The hedgehog gene was first discovered in Drosophila melanogaster, and the pathway is evolutionarily conserved in most animals. Although there are several hedgehog ligands with different protein expression patterns, they share a common plasma membrane receptor, Patched1 and hedgehog signalling pathway activation is transduced through the G-protein-coupled receptor-like protein Smoothened (SMO) and downstream effectors. Functional assays revealed that activation of SMO is dependent on sterol binding, and cholesterol was observed bound to SMO in crystallography experiments. In vertebrates, hedgehog signalling coordinates endochondral ossification and balances osteoblast and osteoclast activation to maintain homeostasis. A recently discovered mutation of SMO in humans (SMOR173C ) is predicted to alter cholesterol binding and is associated with a higher risk of hip OA. Functional studies in mice and human tissue analysis provide evidence that hedgehog signalling is pathologically activated in chondrocytes of osteoarthritic cartilage.
Collapse
Affiliation(s)
- Abbi Elise Smith
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, University of Iceland, Reykjavik, Iceland
| | - Elín Sóley Sigurbjörnsdóttir
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, University of Iceland, Reykjavik, Iceland
| | - Eiríkur Steingrímsson
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, University of Iceland, Reykjavik, Iceland
| | - Sara Sigurbjörnsdóttir
- Faculty of Medicine, Department of Biochemistry and Molecular Biology, University of Iceland, Reykjavik, Iceland.,Faculty of Life and Environmental Sciences, School of Engineering and Natural Sciences, BioMedical Center, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
5
|
Kothandapani A, Jefcoate CR, Jorgensen JS. Cholesterol Contributes to Male Sex Differentiation Through Its Developmental Role in Androgen Synthesis and Hedgehog Signaling. Endocrinology 2021; 162:6204698. [PMID: 33784378 PMCID: PMC8168945 DOI: 10.1210/endocr/bqab066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Indexed: 12/17/2022]
Abstract
Two specialized functions of cholesterol during fetal development include serving as a precursor to androgen synthesis and supporting hedgehog (HH) signaling activity. Androgens are produced by the testes to facilitate masculinization of the fetus. Recent evidence shows that intricate interactions between the HH and androgen signaling pathways are required for optimal male sex differentiation and defects of either can cause birth anomalies indicative of 46,XY male variations of sex development (VSD). Further, perturbations in cholesterol synthesis can cause developmental defects, including VSD, that phenocopy those caused by disrupted androgen or HH signaling, highlighting the functional role of cholesterol in promoting male sex differentiation. In this review, we focus on the role of cholesterol in systemic androgen and local HH signaling events during fetal masculinization and their collective contributions to pediatric VSD.
Collapse
Affiliation(s)
- Anbarasi Kothandapani
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Correspondence: Anbarasi Kothandapani, PhD, Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53705, USA. E-mail:
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705, USA
| | - Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Correspondence: Joan S. Jorgensen, DVM, PhD, Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53705, USA. E-mail:
| |
Collapse
|
6
|
Fleet AJ, Hamel PA. The protein-specific activities of the transmembrane modules of Ptch1 and Ptch2 are determined by their adjacent protein domains. J Biol Chem 2018; 293:16583-16595. [PMID: 30166346 PMCID: PMC6204896 DOI: 10.1074/jbc.ra118.004478] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/17/2018] [Indexed: 01/20/2023] Open
Abstract
Signaling through the Hedgehog (Hh) pathway is mediated by the Patched (Ptch) family of proteins. Although the vertebrate Ptch proteins Ptch1 and Ptch2 harbor two closely related transmembrane modules related to sterol-sensing domains (SSDs), the role of these closely related receptors in the Hh pathway are not equivalent. Ptch1 is essential for development and appears to be the principal receptor mediating responses to Hh ligands, whereas Ptch2 is nonessential, and its role in Hh-signaling remains ambiguous. We hypothesized that the SSDs of the Ptch proteins function as generic modules whose protein-specific activities are determined by the adjacent cytoplasmic and luminal domains. We first showed that individual N-terminal and C-terminal halves of Ptch1 associated noncovalently to mediate ligand-dependent regulation of Hh signaling. The analogous regions of Ptch2 also interacted noncovalently but did not repress the Hh pathway. However, the SSD of Ptch2 were capable of repressing Hh signaling, as determined using chimeric proteins where the SSDs of Ptch1 were replaced by those from Ptch2. Replacement of the SSDs of Ptch1 with the analogous regions from the cholesterol transporter NPC1 failed to produce a chimeric protein capable of Hh repression. Further refinement of the specific regions in Ptch1 and Ptch2 revealed that specific cytoplasmic domains of Ptch1 were necessary but not sufficient for repression of Hh signaling and that the two principal luminal domains of Ptch1 and Ptch2 were interchangeable. These data support a model where the SSDs of the Ptch family proteins exhibit generic activities and that the adjacent cytoplasmic and luminal domains determine their protein-specific activities.
Collapse
Affiliation(s)
- Andrew J Fleet
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Paul A Hamel
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
7
|
Mishra AK, Bernardo-Garcia FJ, Fritsch C, Humberg TH, Egger B, Sprecher SG. Patterning mechanisms diversify neuroepithelial domains in the Drosophila optic placode. PLoS Genet 2018; 14:e1007353. [PMID: 29677185 PMCID: PMC5937791 DOI: 10.1371/journal.pgen.1007353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 05/07/2018] [Accepted: 04/04/2018] [Indexed: 12/31/2022] Open
Abstract
The central nervous system develops from monolayered neuroepithelial sheets. In a first step patterning mechanisms subdivide the seemingly uniform epithelia into domains allowing an increase of neuronal diversity in a tightly controlled spatial and temporal manner. In Drosophila, neuroepithelial patterning of the embryonic optic placode gives rise to the larval eye primordium, consisting of two photoreceptor (PR) precursor types (primary and secondary), as well as the optic lobe primordium, which during larval and pupal stages develops into the prominent optic ganglia. Here, we characterize a genetic network that regulates the balance between larval eye and optic lobe precursors, as well as between primary and secondary PR precursors. In a first step the proneural factor Atonal (Ato) specifies larval eye precursors, while the orphan nuclear receptor Tailless (Tll) is crucial for the specification of optic lobe precursors. The Hedgehog and Notch signaling pathways act upstream of Ato and Tll to coordinate neural precursor specification in a timely manner. The correct spatial placement of the boundary between Ato and Tll in turn is required to control the precise number of primary and secondary PR precursors. In a second step, Notch signaling also controls a binary cell fate decision, thus, acts at the top of a cascade of transcription factor interactions to define PR subtype identity. Our model serves as an example of how combinatorial action of cell extrinsic and cell intrinsic factors control neural tissue patterning.
Collapse
Affiliation(s)
| | | | - Cornelia Fritsch
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Simon G. Sprecher
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- * E-mail:
| |
Collapse
|
8
|
Zarei S, Zarei K, Fritzsch B, Elliott KL. Sonic hedgehog antagonists reduce size and alter patterning of the frog inner ear. Dev Neurobiol 2017; 77:1385-1400. [PMID: 29030893 PMCID: PMC5693645 DOI: 10.1002/dneu.22544] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/28/2017] [Accepted: 10/09/2017] [Indexed: 02/06/2023]
Abstract
Sonic hedgehog (Shh) signaling plays a major role in vertebrate development, from regulation of proliferation to the patterning of various organs. In amniotes, Shh affects dorsoventral patterning in the inner ear but affects anteroposterior patterning in teleost ears. It remains unknown how altered function of Shh relates to morphogenetic changes that coincide with the evolution of limbs and novel auditory organs in the ear. In this study, we used the tetrapod, Xenopus laevis, to test how increasing concentrations of the Shh signal pathway antagonist, Vismodegib, affects ear development. Vismodegib treatment dose dependently alters the development of the ear, hypaxial muscle, and indirectly the Mauthner cell through its interaction with the inner ear afferents. Together, these phenotypes have an effect on escape response. The altered Mauthner cell likely contributes to the increased time to respond to a stimulus. In addition, the increased hypaxial muscle in the trunk likely contributes to the subtle change in animal C-start flexion angle. In the ear, Vismodegib treatment results in decreasing segregation between the gravistatic sensory epithelia as the concentration of Vismodegib increases. Furthermore, at higher doses, there is a loss of the horizontal canal but no enantiomorphic transformation, as in bony fish lacking Shh. Like in amniotes, Shh signaling in frogs affects dorsoventral patterning in the ear, suggesting that auditory sensory evolution in sarcopterygians/tetrapods evolved with a shift of Shh function in axis specification. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1385-1400, 2017.
Collapse
Affiliation(s)
- Sanam Zarei
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
- Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kasra Zarei
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Karen L. Elliott
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
9
|
Deshpande G, Barr J, Gerlitz O, Lebedeva L, Shidlovskii Y, Schedl P. Cells on the move: Modulation of guidance cues during germ cell migration. Fly (Austin) 2017; 11:200-207. [PMID: 28300473 DOI: 10.1080/19336934.2017.1304332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In Drosophila melanogaster the progenitors of the germ-line stem cells, the primordial germ cells (PGCs) are formed on the outside surface of the early embryo, while the somatic gonadal precursor cells (SGPs) are specified during mid-embryogenesis. To form the primitive embryonic gonad, the PGCs travel from outside of the embryo, across the mid-gut and then migrate through the mesoderm to the SGPs. The migratory path of PGCs is dictated by a series of attractive and repulsive cues. Studies in our laboratory have shown that one of the key chemoattractants is the Hedgehog (Hh) ligand. Although, Hh is expressed in other cell types, the long-distance transmission of this ligand is specifically potentiated in the SGPs by the hmgcr isoprenoid biosynthetic pathway. The distant transmission of the Hh ligand is gated by restricting expression of hmgcr to the SGPs. This is particularly relevant in light of the recent findings that an ABC transporter, mdr49 also acts in a mesoderm specific manner to release the germ cell attractant. Our studies have demonstrated that mdr49 functions in hh signaling likely via its role in the transport of cholesterol. Given the importance of cholesterol in the processing and long distance transmission of the Hh ligand, this observation has opened up an exciting avenue concerning the possible role of components of the sterol transport machinery in PGC migration.
Collapse
Affiliation(s)
- Girish Deshpande
- a Department of Molecular Biology , Princeton University , Princeton , NJ , USA
| | - Justinn Barr
- a Department of Molecular Biology , Princeton University , Princeton , NJ , USA
| | - Offer Gerlitz
- b Department of Developmental Biology and Cancer Research, IMRIC , The Hebrew University , Jerusalem , Israel
| | - Lyubov Lebedeva
- c Laboratory of Gene Expression Regulation in Development . Institute of Gene Biology , Moscow , Russia
| | - Yulii Shidlovskii
- c Laboratory of Gene Expression Regulation in Development . Institute of Gene Biology , Moscow , Russia
| | - Paul Schedl
- a Department of Molecular Biology , Princeton University , Princeton , NJ , USA.,c Laboratory of Gene Expression Regulation in Development . Institute of Gene Biology , Moscow , Russia
| |
Collapse
|
10
|
Zhou F, Huang D, Li Y, Hu G, Rao H, Lu Q, Luo S, Wang Y. Nek2A/SuFu feedback loop regulates Gli-mediated Hedgehog signaling pathway. Int J Oncol 2016; 50:373-380. [PMID: 28035348 PMCID: PMC5238777 DOI: 10.3892/ijo.2016.3819] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/09/2016] [Indexed: 12/30/2022] Open
Abstract
Suppressor of Fused (SuFu), one of the most conserved components of the Hedgehog (Hh) signaling, binds Gli transcription factors and impedes activation of target gene expression in mammalian cells. Despite the central importance of SuFu in the Hh pathway, little is known about SuFu regulation. In a previous study, we identified NIMA-related expressed kinase 2A (Nek2A) as a SuFu-interacting protein. Here, we show that Nek2A stabilizes SuFu through impairing ubiquitin/proteasome degradation of SuFu. In addition, Nek2A negatively regulates target genes of Hh signaling as well as Gli2 transcriptional activity. In turn, inhibition of Hh signaling by GANT61 diminishes mRNA and protein levels of Nek2A, and Hh agonist promotes transcription of NEK2A gene. Chromatin immunoprecipitation assays revealed that Gli1 and Gli2 directly bind to the promoter regions of NEK2A gene and induced its transcription. Thus, we uncovered one of the mechanisms by which Nek2A acts as a modulator of the Hh signaling pathway in the context of a novel negative-feedback loop, which may offer new insights into Gli-mediated Hh signaling regulation in development and human diseases.
Collapse
Affiliation(s)
- Fen Zhou
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dengliang Huang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Li
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Guanghui Hu
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hai Rao
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Quqin Lu
- Department of Biostatistics and Epidemiology, School of Public Health, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yao Wang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
11
|
Regulation of Smoothened Trafficking and Hedgehog Signaling by the SUMO Pathway. Dev Cell 2016; 39:438-451. [PMID: 27746045 DOI: 10.1016/j.devcel.2016.09.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 06/14/2016] [Accepted: 09/13/2016] [Indexed: 12/21/2022]
Abstract
Hedgehog (Hh) signaling plays a central role in development and diseases. Hh activates its signal transducer and GPCR-family protein Smoothened (Smo) by inducing Smo phosphorylation, but whether Smo is activated through other post-translational modifications remains unexplored. Here we show that sumoylation acts in parallel with phosphorylation to promote Smo cell-surface expression and Hh signaling. We find that Hh stimulates Smo sumoylation by dissociating it from a desumoylation enzyme Ulp1. Sumoylation of Smo in turn recruits a deubiquitinase UBPY/USP8 to antagonize Smo ubiquitination and degradation, leading to its cell-surface accumulation and elevated Hh pathway activity. We also provide evidence that Shh stimulates sumoylation of mammalian Smo (mSmo) and that sumoylation promotes ciliary localization of mSmo and Shh pathway activity. Our findings reveal a conserved mechanism whereby the SUMO pathway promotes Hh signaling by regulating Smo subcellular localization and shed light on how sumoylation regulates membrane protein trafficking.
Collapse
|
12
|
Buongusto F, Bernardazzi C, Yoshimoto AN, Nanini HF, Coutinho RL, Carneiro AJV, Castelo-Branco MT, de Souza HS. Disruption of the Hedgehog signaling pathway in inflammatory bowel disease fosters chronic intestinal inflammation. Clin Exp Med 2016; 17:351-369. [PMID: 27655445 DOI: 10.1007/s10238-016-0434-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/04/2016] [Indexed: 02/06/2023]
Abstract
Hedgehog (Hh) signaling is essential for intestinal homeostasis and has been associated with inflammation and tissue repair. We hypothesized that Hh signaling could affect the inflammatory process in inflammatory bowel disease (IBD). For this purpose, colon specimens from the inflamed and non-inflamed mucosa of 15 patients with Crohn's disease (CD), 15 with ulcerative colitis, and 15 controls were analyzed by immunohistochemistry and real-time PCR. The production and modulation of cytokines were measured by ELISA from culture explants. Apoptosis was assessed by TUNEL and caspase-3 activity assays. Chemotaxis was evaluated using a transwell system. Primary human intestinal and skin fibroblasts were used for analyzing migration and BrdU incorporation. Hh proteins were generally expressed at the superficial epithelium, and a marked reduction was observed in CD. In the lamina propria, Gli-1 predominantly co-localized with vimentin- and alpha-smooth muscle actin-positive cells, with lower levels observed in CD. In colon explants, Hh stimulation resulted in reduction, while blockade increased, TNF α, IL-17, and TGF β levels. Apoptotic rates were higher in inflamed samples, and they increased after Hh blockade. Levels of Gli-1 mRNA were negatively correlated with caspase-3 activity. Hh blockade increased chemoattraction of monocytes. Primary fibroblasts incorporated more BrdU, but migrated less after Hh blockade. These results suggest that Hh signaling provides a negative feedback to the lamina propria, down-regulating inflammatory cytokines, and inhibiting leukocyte migration and fibroblast proliferation, while favoring fibroblast migration. Therefore, Hh signaling is strongly implicated in the pathogenesis of intestinal inflammation, and it may represent a novel therapeutic target for IBD.
Collapse
Affiliation(s)
- Fernanda Buongusto
- Serviço de Gastroenterologia & Laboratório Multidisciplinar de Pesquisa, Department of Internal Medicine, Hospital Universitário, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 255, Ilha do Fundão, Rio de Janeiro, RJ, 21941-913, Brazil
| | - Claudio Bernardazzi
- Serviço de Gastroenterologia & Laboratório Multidisciplinar de Pesquisa, Department of Internal Medicine, Hospital Universitário, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 255, Ilha do Fundão, Rio de Janeiro, RJ, 21941-913, Brazil
| | - Agnes N Yoshimoto
- Serviço de Gastroenterologia & Laboratório Multidisciplinar de Pesquisa, Department of Internal Medicine, Hospital Universitário, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 255, Ilha do Fundão, Rio de Janeiro, RJ, 21941-913, Brazil
| | - Hayandra F Nanini
- Serviço de Gastroenterologia & Laboratório Multidisciplinar de Pesquisa, Department of Internal Medicine, Hospital Universitário, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 255, Ilha do Fundão, Rio de Janeiro, RJ, 21941-913, Brazil
| | - Raquel L Coutinho
- Serviço de Gastroenterologia & Laboratório Multidisciplinar de Pesquisa, Department of Internal Medicine, Hospital Universitário, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 255, Ilha do Fundão, Rio de Janeiro, RJ, 21941-913, Brazil
| | - Antonio Jose V Carneiro
- Serviço de Gastroenterologia & Laboratório Multidisciplinar de Pesquisa, Department of Internal Medicine, Hospital Universitário, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 255, Ilha do Fundão, Rio de Janeiro, RJ, 21941-913, Brazil
| | - Morgana T Castelo-Branco
- Laboratório de Imunologia Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Heitor S de Souza
- Serviço de Gastroenterologia & Laboratório Multidisciplinar de Pesquisa, Department of Internal Medicine, Hospital Universitário, Universidade Federal do Rio de Janeiro, Rua Prof. Rodolpho Paulo Rocco 255, Ilha do Fundão, Rio de Janeiro, RJ, 21941-913, Brazil. .,D'Or Institute for Research and Education (IDOR), Rio de Janeiro, 22281-100, Brazil.
| |
Collapse
|
13
|
Fleet A, Lee JPY, Tamachi A, Javeed I, Hamel PA. Activities of the Cytoplasmic Domains of Patched-1 Modulate but Are Not Essential for the Regulation of Canonical Hedgehog Signaling. J Biol Chem 2016; 291:17557-68. [PMID: 27325696 DOI: 10.1074/jbc.m116.731745] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Indexed: 12/15/2022] Open
Abstract
The Hedgehog (Hh) pathway is a highly conserved signaling cascade crucial for cell fate determination during embryogenesis. Response to the Hh ligands is mediated by the receptor Patched-1 (Ptch1), a 12-pass transmembrane glycoprotein. Despite its essential role in Hh signaling and its activity as a tumor suppressor, Ptch1 remains largely uncharacterized. We demonstrate here that Ptch1 binds to itself to form oligomeric structures. Oligomerization is mediated by two distinct, structurally disordered, intracellular domains spanning amino acids 584-734 ("middle loop") and 1162-1432 (C terminus). However, oligomerization is not required for Ptch1-dependent regulation of the canonical Hh pathway operating through Smo. Expression of a mutant protein that deletes both regions represses the Hh pathway and responds to the addition of Hh ligand independent of its inability to bind other factors such as Smurf2. Additionally, deletion of the cytoplasmic middle loop domain generates a Ptch1 mutant that, despite binding to Hh ligand, constitutively suppresses Hh signaling and increases the length of primary cilia. Constitutive activity because of deletion of this region is reversed by further deletion of specific sequences in the cytoplasmic C-terminal domain. These data reveal an interaction between the cytoplasmic domains of Ptch1 and that these domains modulate Ptch1 activity but are not essential for regulation of the Hh pathway.
Collapse
Affiliation(s)
- Andrew Fleet
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jennifer P Y Lee
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Aaliya Tamachi
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Imaan Javeed
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Paul A Hamel
- From the Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
14
|
Xavier GM, Seppala M, Barrell W, Birjandi AA, Geoghegan F, Cobourne MT. Hedgehog receptor function during craniofacial development. Dev Biol 2016; 415:198-215. [PMID: 26875496 DOI: 10.1016/j.ydbio.2016.02.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 02/09/2016] [Accepted: 02/10/2016] [Indexed: 01/20/2023]
Abstract
The Hedgehog signalling pathway plays a fundamental role in orchestrating normal craniofacial development in vertebrates. In particular, Sonic hedgehog (Shh) is produced in three key domains during the early formation of the head; neuroectoderm of the ventral forebrain, facial ectoderm and the pharyngeal endoderm; with signal transduction evident in both ectodermal and mesenchymal tissue compartments. Shh signalling from the prechordal plate and ventral midline of the diencephalon is required for appropriate division of the eyefield and forebrain, with mutation in a number of pathway components associated with Holoprosencephaly, a clinically heterogeneous developmental defect characterized by a failure of the early forebrain vesicle to divide into distinct halves. In addition, signalling from the pharyngeal endoderm and facial ectoderm plays an essential role during development of the face, influencing cranial neural crest cells that migrate into the early facial processes. In recent years, the complexity of Shh signalling has been highlighted by the identification of multiple novel proteins that are involved in regulating both the release and reception of this protein. Here, we review the contributions of Shh signalling during early craniofacial development, focusing on Hedgehog receptor function and describing the consequences of disruption for inherited anomalies of this region in both mouse models and human populations.
Collapse
Affiliation(s)
- Guilherme M Xavier
- Department of Craniofacial Development and Stem Cell Biology, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK; Department of Orthodontics, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK
| | - Maisa Seppala
- Department of Craniofacial Development and Stem Cell Biology, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK; Department of Orthodontics, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK
| | - William Barrell
- Department of Craniofacial Development and Stem Cell Biology, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK
| | - Anahid A Birjandi
- Department of Craniofacial Development and Stem Cell Biology, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK
| | - Finn Geoghegan
- Department of Craniofacial Development and Stem Cell Biology, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK
| | - Martyn T Cobourne
- Department of Craniofacial Development and Stem Cell Biology, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK; Department of Orthodontics, King's College London Dental Institute, Floor 27, Guy's Hospital, London SE1 9RT, UK.
| |
Collapse
|
15
|
Klafke R, Prem Anand AA, Wurst W, Prakash N, Wizenmann A. Differences in the spatiotemporal expression and epistatic gene regulation of the mesodiencephalic dopaminergic precursor marker PITX3 during chicken and mouse development. Development 2016; 143:691-702. [DOI: 10.1242/dev.126748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 01/05/2016] [Indexed: 11/20/2022]
Abstract
Mesodiencephalic dopaminergic (mdDA) neurons are located in the ventral mesencephalon and caudal diencephalon of all tetrapod species studied so far. They are the most prominent DA neuronal population and are implicated in control and modulation of motor, cognitive and rewarding/affective behaviors. Their degeneration or dysfunction is intimately linked to several neurological and neuropsychiatric human diseases. To gain further insights into their generation, we studied spatiotemporal expression patterns and epistatic interactions in chick embryos of selected marker genes and signaling pathways associated with mdDA neuron development in mouse. We detected striking differences in the expression patterns of the chick orthologs of the mouse mdDA marker genes Pitx3 and Aldh1a1, which suggests important differences between the species in the generation/generating of these cells. We also discovered that the Sonic hedgehog signaling pathway is both, necessary and sufficient for the induction of ectopic PITX3 expression in chick mesencephalon downstream of WNT9A induced LMX1a transcription. These aspects of early chicken development resemble the ontogeny of zebrafish diencephalic DA neuronal populations, and suggest a divergence between birds and mammals during evolution.
Collapse
Affiliation(s)
- Ruth Klafke
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - A. Alwin Prem Anand
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Oesterbergstrasse 3, 72074 Tuebingen, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Standort München, Schillerstr. 44, 80336 München, Germany
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Schillerstrasse 44, 80336 München, Germany
| | - Nilima Prakash
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Andrea Wizenmann
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Oesterbergstrasse 3, 72074 Tuebingen, Germany
| |
Collapse
|
16
|
Callahan BP, Wang C. Hedgehog Cholesterolysis: Specialized Gatekeeper to Oncogenic Signaling. Cancers (Basel) 2015; 7:2037-53. [PMID: 26473928 PMCID: PMC4695875 DOI: 10.3390/cancers7040875] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/22/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022] Open
Abstract
Discussions of therapeutic suppression of hedgehog (Hh) signaling almost exclusively focus on receptor antagonism; however, hedgehog's biosynthesis represents a unique and potentially targetable aspect of this oncogenic signaling pathway. Here, we review a key biosynthetic step called cholesterolysis from the perspectives of structure/function and small molecule inhibition. Cholesterolysis, also called cholesteroylation, generates cholesterol-modified Hh ligand via autoprocessing of a hedgehog precursor protein. Post-translational modification by cholesterol appears to be restricted to proteins in the hedgehog family. The transformation is essential for Hh biological activity and upstream of signaling events. Despite its decisive role in generating ligand, cholesterolysis remains conspicuously unexplored as a therapeutic target.
Collapse
Affiliation(s)
- Brian P Callahan
- Chemistry Department, Binghamton University 4400 Vestal Parkway East, Binghamton, NY 13902, USA.
| | - Chunyu Wang
- Biology Department, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA.
| |
Collapse
|
17
|
Arensdorf AM, Marada S, Ogden SK. Smoothened Regulation: A Tale of Two Signals. Trends Pharmacol Sci 2015; 37:62-72. [PMID: 26432668 DOI: 10.1016/j.tips.2015.09.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 02/09/2023]
Abstract
The G protein-coupled receptor (GPCR) Smoothened (Smo) is the signal transducer of the developmentally and therapeutically relevant Hedgehog (Hh) pathway. Although recent structural analyses have advanced our understanding of Smo biology, several questions remain. Chief among them are the identity of its natural ligand, the regulatory processes controlling its activation, and the mechanisms by which it signals to downstream effectors. In this review, we discuss recent discoveries from multiple model systems that have set the stage for solving these mysteries. We focus on the roles of distinct Smo functional domains, post-translational modifications, and trafficking, and conclude by discussing their contributions to signal output.
Collapse
Affiliation(s)
- Angela M Arensdorf
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, 262 Danny Thomas Place MS#340, Memphis, TN 38105, USA
| | - Suresh Marada
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, 262 Danny Thomas Place MS#340, Memphis, TN 38105, USA
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, 262 Danny Thomas Place MS#340, Memphis, TN 38105, USA.
| |
Collapse
|
18
|
Tian A, Shi Q, Jiang A, Li S, Wang B, Jiang J. Injury-stimulated Hedgehog signaling promotes regenerative proliferation of Drosophila intestinal stem cells. ACTA ACUST UNITED AC 2015; 208:807-19. [PMID: 25753035 PMCID: PMC4362464 DOI: 10.1083/jcb.201409025] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In response to injury, Hedgehog signaling regulates the production of Upd2 in enteroblasts, which in turn activates the JAK–STAT pathway to drive intestinal stem cell proliferation. Many adult tissues are maintained by resident stem cells that elevate their proliferation in response to injury. The regulatory mechanisms underlying regenerative proliferation are still poorly understood. Here we show that injury induces Hedgehog (Hh) signaling in enteroblasts (EBs) to promote intestinal stem cell (ISC) proliferation in Drosophila melanogaster adult midgut. Elevated Hh signaling by patched (ptc) mutations drove ISC proliferation noncell autonomously. Inhibition of Hh signaling in the ISC lineage compromised injury-induced ISC proliferation but had little if any effect on homeostatic proliferation. Hh signaling acted in EBs to regulate the production of Upd2, which activated the JAK–STAT pathway to promote ISC proliferation. Furthermore, we show that Hh signaling is stimulated by DSS through the JNK pathway and that inhibition of Hh signaling in EBs prevented DSS-stimulated ISC proliferation. Hence, our study uncovers a JNK–Hh–JAK–STAT signaling axis in the regulation of regenerative stem cell proliferation.
Collapse
Affiliation(s)
- Aiguo Tian
- Department of Developmental Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390
| | - Qing Shi
- Department of Developmental Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390
| | - Alice Jiang
- Department of Developmental Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390
| | - Shuangxi Li
- Department of Developmental Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390
| | - Bing Wang
- Department of Developmental Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390
| | - Jin Jiang
- Department of Developmental Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390 Department of Developmental Biology and Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390
| |
Collapse
|
19
|
Camp D, Haitian He B, Li S, Althaus IW, Holtz AM, Allen BL, Charron F, van Meyel DJ. Ihog and Boi elicit Hh signaling via Ptc but do not aid Ptc in sequestering the Hh ligand. Development 2014; 141:3879-88. [PMID: 25231763 DOI: 10.1242/dev.103564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hedgehog (Hh) proteins are secreted molecules essential for tissue development in vertebrates and invertebrates. Hh reception via the 12-pass transmembrane protein Patched (Ptc) elicits intracellular signaling through Smoothened (Smo). Hh binding to Ptc is also proposed to sequester the ligand, limiting its spatial range of activity. In Drosophila, Interference hedgehog (Ihog) and Brother of ihog (Boi) are two conserved and redundant transmembrane proteins that are essential for Hh pathway activation. How Ihog and Boi activate signaling in response to Hh remains unknown; each can bind both Hh and Ptc and so it has been proposed that they are essential for both Hh reception and sequestration. Using genetic epistasis we established here that Ihog and Boi, and their orthologs in mice, act upstream or at the level of Ptc to allow Hh signal transduction. In the Drosophila developing wing model we found that it is through Hh pathway activation that Ihog and Boi maintain the boundary between the anterior and posterior compartments. We dissociated the contributions of Ptc from those of Ihog/Boi and, surprisingly, found that cells expressing Ptc can retain and sequester the Hh ligand without Ihog and Boi, but that Ihog and Boi cannot do so without Ptc. Together, these results reinforce the central role for Ptc in Hh binding in vivo and demonstrate that, although Ihog and Boi are dispensable for Hh sequestration, they are essential for pathway activation because they allow Hh to inhibit Ptc and thereby relieve its repression of Smo.
Collapse
Affiliation(s)
- Darius Camp
- McGill Centre for Research in Neuroscience and the McGill University Health Centre Research Institute, 1650 Cedar Avenue, Montreal, Quebec, Canada H3G 1A4 Molecular Biology of Neural Development, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec, Canada H2W 1R7 Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada H3A 1A3
| | - Billy Haitian He
- McGill Centre for Research in Neuroscience and the McGill University Health Centre Research Institute, 1650 Cedar Avenue, Montreal, Quebec, Canada H3G 1A4
| | - Sally Li
- McGill Centre for Research in Neuroscience and the McGill University Health Centre Research Institute, 1650 Cedar Avenue, Montreal, Quebec, Canada H3G 1A4
| | - Irene W Althaus
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexander M Holtz
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Frédéric Charron
- Molecular Biology of Neural Development, Institut de Recherches Cliniques de Montréal (IRCM), Montreal, Quebec, Canada H2W 1R7 Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada H3A 1A3 Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4 Program in Neuroengineering, McGill University, Montreal, Quebec, Canada H3A 2K6 Department of Medicine, University of Montreal, Montreal, Quebec, Canada H3T 1J4 Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada H3A 0C7 Department of Biology, McGill University, Montreal, Quebec, Canada H3A 1B1
| | - Donald J van Meyel
- McGill Centre for Research in Neuroscience and the McGill University Health Centre Research Institute, 1650 Cedar Avenue, Montreal, Quebec, Canada H3G 1A4 Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada H3A 1A3 Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada H3A 2B4 Department of Biology, McGill University, Montreal, Quebec, Canada H3A 1B1
| |
Collapse
|
20
|
Li S, Ma G, Wang B, Jiang J. Hedgehog induces formation of PKA-Smoothened complexes to promote Smoothened phosphorylation and pathway activation. Sci Signal 2014; 7:ra62. [PMID: 24985345 PMCID: PMC4621970 DOI: 10.1126/scisignal.2005414] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hedgehog (Hh) is a secreted glycoprotein that binds its receptor Patched to activate the G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor-like protein Smoothened (Smo). In Drosophila, protein kinase A (PKA) phosphorylates and activates Smo in cells stimulated with Hh. In unstimulated cells, PKA phosphorylates and inhibits the transcription factor Cubitus interruptus (Ci). We found that in cells exposed to Hh, the catalytic subunit of PKA (PKAc) bound to the juxtamembrane region of the carboxyl terminus of Smo. PKA-mediated phosphorylation of Smo further enhanced its association with PKAc to form stable kinase-substrate complexes that promoted the PKA-mediated transphosphorylation of Smo dimers. We identified multiple basic residues in the carboxyl terminus of Smo that were required for interaction with PKAc, Smo phosphorylation, and Hh pathway activation. Hh induced a switch from the association of PKAc with a cytosolic complex of Ci and the kinesin-like protein Costal2 (Cos2) to a membrane-bound Smo-Cos2 complex. Thus, our study uncovers a previously uncharacterized mechanism for regulation of PKA activity and demonstrates that the signal-regulated formation of kinase-substrate complexes plays a central role in Hh signal transduction.
Collapse
Affiliation(s)
- Shuang Li
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Guoqiang Ma
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Bing Wang
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Jin Jiang
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA. Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA.
| |
Collapse
|
21
|
Harvey MC, Fleet A, Okolowsky N, Hamel PA. Distinct effects of the mesenchymal dysplasia gene variant of murine Patched-1 protein on canonical and non-canonical Hedgehog signaling pathways. J Biol Chem 2014; 289:10939-10949. [PMID: 24570001 DOI: 10.1074/jbc.m113.514844] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Hedgehog (Hh) signaling requires regulation of the receptor Patched-1 (Ptch1), which, in turn, regulates Smoothened activity (canonical Hh signaling) as well as other non-canonical signaling pathways. The mutant Ptch1 allele mesenchymal dysplasia (mes), which truncates the Ptch1 C terminus, produces a limited spectrum of developmental defects in mice as well as deregulation of canonical Hh signaling in some, but not all, affected tissues. Paradoxically, mes suppresses canonical Hh signaling and binds to Hh ligands with an affinity similar to wild-type mouse Ptch1 (mPtch1). We characterized the distinct activities of the mes variant of mPtch1 mediating Hh signaling through both canonical and non-canonical pathways. We demonstrated that mPtch1 bound c-src in an Hh-regulated manner. Stimulation with Sonic Hedgehog (Shh) of primary mammary mesenchymal cells from wild-type and mes animals activated Erk1/2. Although Shh activated c-src in wild-type cells, c-src was constitutively activated in mes mesenchymal cells. Transient assays showed that wild-type mPtch1, mes, or mPtch1 lacking the C terminus repressed Hh signaling in Ptch1-deficient mouse embryo fibroblasts and that repression was reversed by Shh, revealing that the C terminus was dispensable for mPtch1-dependent regulation of canonical Hh signaling. In contrast to these transient assays, constitutively high levels of mGli1 but not mPtch1 were present in primary mammary mesenchymal cells from mes mice, whereas the expression of mPtch1 was similarly induced in both mes and wild-type cells. These data define a novel signal transduction pathway involving c-src that is activated by the Hh ligands and reveals the requirement for the C terminus of Ptch in regulation of canonical and non-canonical Hh signaling pathways.
Collapse
Affiliation(s)
- Malcolm C Harvey
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Andrew Fleet
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Nadia Okolowsky
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Paul A Hamel
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
22
|
Hartl TA, Scott MP. Wing tips: The wing disc as a platform for studying Hedgehog signaling. Methods 2014; 68:199-206. [PMID: 24556557 DOI: 10.1016/j.ymeth.2014.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/03/2014] [Accepted: 02/06/2014] [Indexed: 12/26/2022] Open
Abstract
Hedgehog (Hh) signal transduction is necessary for the development of most mammalian tissues and can go awry and cause birth defects or cancer. Hh signaling was initially described in Drosophila, and much of what we know today about mammalian Hh signaling was directly guided by discoveries in the fly. Indeed, Hh signaling is a wonderful example of the use of non-vertebrate model organisms to make basic discoveries that lead to new disease treatment. The first pharmaceutical to treat hyperactive Hh signaling in Basal Cell Carcinoma was released in 2012, approximately 30 years after the isolation of Hh mutants in Drosophila. The study of Hh signaling has been greatly facilitated by the imaginal wing disc, a tissue with terrific experimental advantages. Studies using the wing disc have led to an understanding of Hh ligand processing, packaging into particles for transmission, secretion, reception, signal transduction, target gene activation, and tissue patterning. Here we describe the imaginal wing disc, how Hh patterns this tissue, and provide methods to use wing discs to study Hh signaling in Drosophila. The tools and approaches we highlight form the cornerstone of research efforts in many laboratories that use Drosophila to study Hh signaling, and are essential for ongoing discoveries.
Collapse
Affiliation(s)
- Tom A Hartl
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew P Scott
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
23
|
Huang S, Zhang Z, Zhang C, Lv X, Zheng X, Chen Z, Sun L, Wang H, Zhu Y, Zhang J, Yang S, Lu Y, Sun Q, Tao Y, Liu F, Zhao Y, Chen D. Activation of Smurf E3 ligase promoted by smoothened regulates hedgehog signaling through targeting patched turnover. PLoS Biol 2013; 11:e1001721. [PMID: 24302888 PMCID: PMC3841102 DOI: 10.1371/journal.pbio.1001721] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 10/18/2013] [Indexed: 12/26/2022] Open
Abstract
Hedgehog signaling plays conserved roles in controlling embryonic development; its dysregulation has been implicated in many human diseases including cancers. Hedgehog signaling has an unusual reception system consisting of two transmembrane proteins, Patched receptor and Smoothened signal transducer. Although activation of Smoothened and its downstream signal transduction have been intensively studied, less is known about how Patched receptor is regulated, and particularly how this regulation contributes to appropriate Hedgehog signal transduction. Here we identified a novel role of Smurf E3 ligase in regulating Hedgehog signaling by controlling Patched ubiquitination and turnover. Moreover, we showed that Smurf-mediated Patched ubiquitination depends on Smo activity in wing discs. Mechanistically, we found that Smo interacts with Smurf and promotes it to mediate Patched ubiquitination by targeting the K1261 site in Ptc. The further mathematic modeling analysis reveals that a bidirectional control of activation of Smo involving Smurf and Patched is important for signal-receiving cells to precisely interpret external signals, thereby maintaining Hedgehog signaling reliability. Finally, our data revealed an evolutionarily conserved role of Smurf proteins in controlling Hh signaling by targeting Ptc during development.
Collapse
Affiliation(s)
- Shoujun Huang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhao Zhang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chunxia Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Beijing, China
| | - Xiangdong Lv
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiudeng Zheng
- Centre for Computational and Evolutionary Biology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Animal Ecology, Conservational Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhenping Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Liwei Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Beijing, China
| | - Hailong Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yuanxiang Zhu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jing Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Beijing, China
| | - Shuyan Yang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yi Lu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qinmiao Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Beijing, China
| | - Yi Tao
- Key Laboratory of Animal Ecology, Conservational Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Feng Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Chinese Academy of Sciences, Beijing, China
| | - Yun Zhao
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dahua Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
24
|
Myers BR, Sever N, Chong YC, Kim J, Belani JD, Rychnovsky S, Bazan JF, Beachy PA. Hedgehog pathway modulation by multiple lipid binding sites on the smoothened effector of signal response. Dev Cell 2013; 26:346-57. [PMID: 23954590 DOI: 10.1016/j.devcel.2013.07.015] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/17/2013] [Accepted: 07/18/2013] [Indexed: 01/06/2023]
Abstract
Hedgehog (Hh) signaling during development and in postembryonic tissues requires activation of the 7TM oncoprotein Smoothened (Smo) by mechanisms that may involve endogenous lipidic modulators. Exogenous Smo ligands previously identified include the plant sterol cyclopamine (and its therapeutically useful synthetic mimics) and hydroxylated cholesterol derivatives (oxysterols); Smo is also highly sensitive to cellular sterol levels. The relationships between these effects are unclear because the relevant Smo structural determinants are unknown. We identify the conserved extracellular cysteine-rich domain (CRD) as the site of action for oxysterols on Smo, involving residues structurally analogous to those contacting the Wnt lipid adduct in the homologous Frizzled CRD; this modulatory effect is distinct from that of cyclopamine mimics, from Hh-mediated regulation, and from the permissive action of cellular sterol pools. These results imply that Hh pathway activity is sensitive to lipid binding at several Smo sites, suggesting mechanisms for tuning by multiple physiological inputs.
Collapse
Affiliation(s)
- Benjamin R Myers
- Department of Biochemistry, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Villani RM, Waters MJ, Wainwright BJ. Murine basal cell carcinoma leads to tumor-mediated alterations in endocrine Igf1 signaling. Endocr Relat Cancer 2013; 20:273-81. [PMID: 23404854 DOI: 10.1530/erc-12-0307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The intrinsic properties underlying cancer development are extensively studied while the effect of a cancer on the host is often overlooked. Activation of the Hedgehog (Hh) signaling pathway underlies a number of types of common human cancers, yet little is known concerning endocrine signaling in such tumors. Here, we investigated endocrine signaling in a murine model of basal cell carcinoma (BCC) of the skin, the most common cancer. BCCs were generated by the activation of Hh signaling resulting from the specific deletion of the Ptch1 gene in the developing epidermis. Subsequently, a severe growth deficiency was observed in the murine BCC model, and we identified a deficiency of circulating IGF1 (Igf1). We demonstrate that Hh pathway activation in murine BCC induces IGF binding proteins, thereby regulating Igf1 sequestration into the skin and skewing Igf endocrine signaling. Significantly, these results show that Hh-induced tumors can have endocrine effects on normal tissues that in turn can greatly impact the host. This study not only identifies that Igf is important in Hh-associated skin tumors but also exemplifies the need to consider endocrine signaling when interpreting complex in vivo tumor models.
Collapse
Affiliation(s)
- Rehan M Villani
- Institute for Molecular Bioscience, University of Queensland, 360 Carmody Road, St Lucia, Brisbane, Queensland 4072, Australia
| | | | | |
Collapse
|
26
|
Baker NE. Developmental regulation of nucleolus size during Drosophila eye differentiation. PLoS One 2013; 8:e58266. [PMID: 23472166 PMCID: PMC3589261 DOI: 10.1371/journal.pone.0058266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 02/05/2013] [Indexed: 11/29/2022] Open
Abstract
When cell cycle withdrawal accompanies terminal differentiation, biosynthesis and cellular growth are likely to change also. In this study, nucleolus size was monitored during cell fate specification in the Drosophila eye imaginal disc using fibrillarin antibody labeling. Nucleolus size is an indicator of ribosome biogenesis and can correlate with cellular growth rate. Nucleolar size was reduced significantly during cell fate specification and differentiation, predominantly as eye disc cells entered a cell cycle arrest that preceded cell fate specification. This reduction in nucleolus size required Dpp and Hh signaling. A transient enlargement of the nucleolus accompanied cell division in the Second Mitotic Wave. Nucleoli continued to diminish in postmitotic cells following fate specification. These results suggest that cellular growth is regulated early in the transition from proliferating progenitor cells to terminal cell fate specification, contemporary with regulation of the cell cycle, and requiring the same extracellular signals.
Collapse
Affiliation(s)
- Nicholas E Baker
- Departments of Genetics, Ophthalmology and Visual Sciences, and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
27
|
Avanesov A, Blair SS. The Drosophila WIF1 homolog Shifted maintains glypican-independent Hedgehog signaling and interacts with the Hedgehog co-receptors Ihog and Boi. Development 2012; 140:107-16. [PMID: 23154411 DOI: 10.1242/dev.078444] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hedgehog (Hh) family proteins are secreted signaling ligands whose short- and long-range activities transform cellular fates in multiple contexts in organisms ranging from metazoans to humans. In the developing Drosophila wing, extracellular Hh binds to cell-bound glypican heparan sulfate proteoglycans (HSPGs) and the secreted protein Shifted (Shf), a member of Wnt inhibitory factor 1 (WIF1) family. The glypicans and Shf are required for long-range Hh movement and signaling; it has been proposed that Shf promotes long-range Hh signaling by reinforcing binding between Hh and the glypicans, and that much or all of glypican function in Hh signaling requires Shf. However, we will show here that Shf maintains short-range Hh signaling in the wing via a mechanism that does not require the presence of or binding to the Drosophila glypicans Dally and Dally-like protein. Conversely, we demonstrate interactions between Hh and the glypicans that are maintained, and even strengthened, in the absence of Shf. We present evidence that Shf binds to the CDO/BOC family Hh co-receptors Interference hedgehog (Ihog) and Brother of Ihog, suggesting that Shf regulates short-range Hh signaling through interactions with the receptor complex. In support of a functional interaction between Ihog and members of the Shf/WIF1 family, we show that Ihog can increase the Wnt-inhibitory activity of vertebrate WIF1; this result raises the possibility of interactions between WIF1 and vertebrate CDO/BOC family members.
Collapse
Affiliation(s)
- Andrei Avanesov
- Department of Zoology, University of Wisconsin, Madison, WI 53706, USA
| | | |
Collapse
|
28
|
Michel M, Kupinski AP, Raabe I, Bökel C. Hh signalling is essential for somatic stem cell maintenance in the Drosophila testis niche. Development 2012; 139:2663-9. [PMID: 22745310 DOI: 10.1242/dev.075242] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the Drosophila testis, germline stem cells (GSCs) and somatic cyst stem cells (CySCs) are arranged around a group of postmitotic somatic cells, termed the hub, which produce a variety of growth factors contributing to the niche microenvironment that regulates both stem cell pools. Here we show that CySC but not GSC maintenance requires Hedgehog (Hh) signalling in addition to Jak/Stat pathway activation. CySC clones unable to transduce the Hh signal are lost by differentiation, whereas pathway overactivation leads to an increase in proliferation. However, unlike cells ectopically overexpressing Jak/Stat targets, the additional cells generated by excessive Hh signalling remain confined to the testis tip and retain the ability to differentiate. Interestingly, Hh signalling also controls somatic cell populations in the fly ovary and the mammalian testis. Our observations might therefore point towards a higher degree of organisational homology between the somatic components of gonads across the sexes and phyla than previously appreciated.
Collapse
|
29
|
A network of broadly expressed HLH genes regulates tissue-specific cell fates. Cell 2012; 147:881-92. [PMID: 22078884 DOI: 10.1016/j.cell.2011.08.055] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 07/06/2011] [Accepted: 08/17/2011] [Indexed: 12/11/2022]
Abstract
Spatial and temporal expression of specific basic-helix-loop-helix (bHLH) transcription factors defines many types of cellular differentiation. We find that a distinct mechanism regulates the much broader expression of the heterodimer partners of these specific factors and impinges on differentiation. In Drosophila, a cross-interacting regulatory network links expression of the E protein Daughterless (Da), which heterodimerizes with bHLH proteins to activate them, with expression of the Id protein Extramacrochaetae (Emc), which antagonizes bHLH proteins. Coupled transcriptional feedback loops maintain the widespread Emc expression that restrains Da expression, opposing bHLH-dependent differentiation while enhancing growth and cell survival. Where extracellular signals repress emc, Da expression can increase. This defines regions of proneural ectoderm independently from the proneural bHLH genes. Similar regulation is found in multiple Drosophila tissues and in mammalian cells and therefore is likely to be a conserved general feature of developmental regulation by HLH proteins.
Collapse
|
30
|
Abstract
The Hedgehog (Hh) pathway is a conserved signalling system essential for embryonic development and for the maintenance of self-renewal pathways in progenitor cells. Mutations that deregulate Hh signalling are directly implicated in basal cell carcinoma and medulloblastoma. The mechanisms of Hh pathway activation in cancers in which no pathway mutations have been identified are less clear, but of great translational significance. Small molecule inhibitors of the pathway, many of which are in early phase clinical trials, may shed further light on this question. Canonical Hh signalling promotes the expression of target genes through the Glioma-associated oncogene (GLI) transcription factors. There is now increasing evidence suggesting that 'non-canonical' Hh signalling mechanisms, some of which are independent of GLI-mediated transcription, may be important in cancer and development. The focus of this review is to summarise some of the known mechanisms of Hh signalling as well as its emerging role in cancer.
Collapse
Affiliation(s)
- Kieren D Marini
- Monash Institute of Medical Research, Centre for Cancer Research, Monash University, Victoria, Australia
| | | | | | | |
Collapse
|
31
|
Shi Q, Li S, Jia J, Jiang J. The Hedgehog-induced Smoothened conformational switch assembles a signaling complex that activates Fused by promoting its dimerization and phosphorylation. Development 2011; 138:4219-31. [PMID: 21852395 DOI: 10.1242/dev.067959] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hedgehog (Hh) transduces signal by regulating the subcellular localization and conformational state of the GPCR-like protein Smoothened (Smo) but how Smo relays the signal to cytoplasmic signaling components remains poorly understood. Here, we show that Hh-induced Smo conformational change recruits Costal2 (Cos2)/Fused (Fu) and promotes Fu kinase domain dimerization. We find that induced dimerization through the Fu kinase domain activates Fu by inducing multi-site phosphorylation of its activation loop (AL) and phospho-mimetic mutations of AL activate the Hh pathway. Interestingly, we observe that graded Hh signals progressively increase Fu kinase domain dimerization and AL phosphorylation, suggesting that Hh activates Fu in a dose-dependent manner. Moreover, we find that activated Fu regulates Cubitus interruptus (Ci) by both promoting its transcriptional activator activity and inhibiting its proteolysis into a repressor form. We provide evidence that activated Fu exerts these regulations by interfering with the formation of Ci-Sufu and Ci-Cos2-kinase complexes that normally inhibit Ci activity and promote its processing. Taken together, our results suggest that Hh-induced Smo conformational change facilitates the assembly of active Smo-Cos2-Fu signaling complexes that promote Fu kinase domain dimerization, phosphorylation and activation, and that Fu regulates both the activator and repressor forms of Ci.
Collapse
Affiliation(s)
- Qing Shi
- Department of Developmental Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | | | | | | |
Collapse
|
32
|
Schulte G. International Union of Basic and Clinical Pharmacology. LXXX. The class Frizzled receptors. Pharmacol Rev 2011; 62:632-67. [PMID: 21079039 DOI: 10.1124/pr.110.002931] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The receptor class Frizzled, which has recently been categorized as a separate group of G protein-coupled receptors by the International Union of Basic and Clinical Pharmacology, consists of 10 Frizzleds (FZD(1-10)) and Smoothened (SMO). The FZDs are activated by secreted lipoglycoproteins of the Wingless/Int-1 (WNT) family, whereas SMO is indirectly activated by the Hedgehog (HH) family of proteins acting on the transmembrane protein Patched (PTCH). Recent years have seen major advances in our knowledge about these seven-transmembrane-spanning proteins, including: receptor function, molecular mechanisms of signal transduction, and the receptor's role in embryonic patterning, physiology, cancer, and other diseases. Despite intense efforts, many question marks and challenges remain in mapping receptor-ligand interaction, signaling routes, mechanisms of specificity and how these molecular details underlie disease and also the receptor's important role in physiology. This review therefore focuses on the molecular aspects of WNT/FZD and HH/SMO signaling discussing receptor structure, mechanisms of signal transduction, accessory proteins, receptor dynamics, and the possibility of targeting these signaling pathways pharmacologically.
Collapse
Affiliation(s)
- Gunnar Schulte
- Section of Receptor Biology & Signaling, Dept. of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Nicholson SC, Nicolay BN, Frolov MV, Moberg KH. Notch-dependent expression of the archipelago ubiquitin ligase subunit in the Drosophila eye. Development 2010; 138:251-60. [PMID: 21148181 DOI: 10.1242/dev.054429] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
archipelago (ago)/Fbw7 encodes a conserved protein that functions as the substrate-receptor component of a polyubiquitin ligase that suppresses tissue growth in flies and tumorigenesis in vertebrates. Ago/Fbw7 targets multiple proteins for degradation, including the G1-S regulator Cyclin E and the oncoprotein dMyc/c-Myc. Despite prominent roles in growth control, little is known about the signals that regulate Ago/Fbw7 abundance in developing tissues. Here we use the Drosophila eye as a model to identify developmental signals that regulate ago expression. We find that expression of ago mRNA and protein is induced by passage of the morphogenetic furrow (MF) and identify the hedgehog (hh) and Notch (N) pathways as elements of this inductive mechanism. Cells mutant for N pathway components, or hh-defective cells that express reduced levels of the Notch ligand Delta, fail to upregulate ago transcription in the region of the MF; reciprocally, ectopic N activation in eye discs induces expression of ago mRNA. A fragment of the ago promoter that contains consensus binding sites for the N pathway transcription factor Su(H) is bound by Su(H) and confers N-inducibility in cultured cells. The failure to upregulate ago in N pathway mutant cells correlates with accumulation of the SCF-Ago target Cyclin E in the area of the MF, and this is rescued by re-expression of ago. These data suggest a model in which N acts through ago to restrict levels of the pro-mitotic factor Cyclin E. This N-Ago-Cyclin E link represents a significant new cell cycle regulatory mechanism in the developing eye.
Collapse
Affiliation(s)
- Sarah C Nicholson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
34
|
Hartman TR, Zinshteyn D, Schofield HK, Nicolas E, Okada A, O'Reilly AM. Drosophila Boi limits Hedgehog levels to suppress follicle stem cell proliferation. ACTA ACUST UNITED AC 2010; 191:943-52. [PMID: 21098113 PMCID: PMC2995164 DOI: 10.1083/jcb.201007142] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The Boi receptor regulates stem cell function by sequestering the diffusible hedgehog ligand. Stem cells depend on signals from cells within their microenvironment, or niche, as well as factors secreted by distant cells to regulate their maintenance and function. Here we show that Boi, a Hedgehog (Hh)-binding protein, is a novel suppressor of proliferation of follicle stem cells (FSCs) in the Drosophila ovary. Hh is expressed in apical cells, distant from the FSC niche, and diffuses to reach FSCs, where it promotes FSC proliferation. We show that Boi is expressed in apical cells and exerts its suppressive effect on FSC proliferation by binding to and sequestering Hh on the apical cell surface, thereby inhibiting Hh diffusion. Our studies demonstrate that cells distant from the local niche can regulate stem cell function through ligand sequestration, a mechanism that likely is conserved in other epithelial tissues.
Collapse
|
35
|
Abstract
In vertebrate hedgehog signaling, hedgehog ligands are processed to become bilipidated and then multimerize, which allows them to leave the signaling cell via Dispatched 1 and become transported via glypicans and megalin to the responding cells. Hedgehog then interacts with a complex of Patched 1 and Cdo/Boc, which activates endocytic Smoothened to the cilium. Patched 1 regulates the activity of Smoothened (1) via Vitamin D3, which inhibits Smoothened in the absence of hedgehog ligand or (2) via oxysterols, which activate Smoothened in the presence of hedgehog ligand. Hedgehog ligands also interact with Hip1, Patched 2, and Gas1, which regulate the range as well as the level of hedgehog signaling. In vertebrates, Smoothened is shortened at its C-terminal end and lacks most of the phosphorylation sites of importance in Drosophila. Cos2, also of importance in Drosophila, plays no role in mammalian transduction, nor do its homologs Kif7 and Kif27. The cilium may provide a function analogous to that of Cos2 by linking Smoothened to the modulation of Gli transcription factors. Disorders associated with the hedgehog signaling network follow, including nevoid basal cell carcinoma syndrome, holoprosencephaly, Smith-Lemli-Opitz syndrome, Greig cephalopolysyndactyly syndrome, Pallister-Hall syndrome, Carpenter syndrome, and Rubinstein-Taybi syndrome.
Collapse
Affiliation(s)
- M Michael Cohen
- Department of Oral & Maxillofacial Sciences, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
36
|
Chou YH, Zheng X, Beachy PA, Luo L. Patterning axon targeting of olfactory receptor neurons by coupled hedgehog signaling at two distinct steps. Cell 2010; 142:954-66. [PMID: 20850015 DOI: 10.1016/j.cell.2010.08.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 05/23/2010] [Accepted: 07/26/2010] [Indexed: 02/04/2023]
Abstract
We present evidence for a coupled two-step action of Hedgehog signaling in patterning axon targeting of Drosophila olfactory receptor neurons (ORNs). In the first step, differential Hedgehog pathway activity in peripheral sensory organ precursors creates ORN populations with different levels of the Patched receptor. Different Patched levels in ORNs then determine axonal responsiveness to target-derived Hedgehog in the brain: only ORN axons that do not express high levels of Patched are responsive to and require a second step of Hedgehog signaling for target selection. Hedgehog signaling in the imaginal sensory organ precursors thus confers differential ORN responsiveness to Hedgehog-mediated axon targeting in the brain. This mechanism contributes to the spatial coordination of ORN cell bodies in the periphery and their glomerular targets in the brain. Such coupled two-step signaling may be more generally used to coordinate other spatially and temporally segregated developmental events.
Collapse
Affiliation(s)
- Ya-Hui Chou
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
37
|
Jia H, Liu Y, Xia R, Tong C, Yue T, Jiang J, Jia J. Casein kinase 2 promotes Hedgehog signaling by regulating both smoothened and Cubitus interruptus. J Biol Chem 2010; 285:37218-26. [PMID: 20876583 DOI: 10.1074/jbc.m110.174565] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Casein kinase 2 (CK2) is a typical serine/threonine kinase consisting of α and β subunits and has been implicated in many cellular and developmental processes. In this study, we demonstrate that CK2 is a positive regulator of the Hedgehog (Hh) signal transduction pathway. We found that inactivation of CK2 by CK2β RNAi enhances the loss-of-Hh wing phenotype induced by a dominant negative form of Smoothened (Smo). CK2β RNAi attenuates Hh-induced Smo accumulation and down-regulates Hh target gene expression, whereas increasing CK2 activity by coexpressing CK2α and CK2β increases Smo accumulation and induces ectopic Hh target gene expression. We identified the serine residues in Smo that can be phosphorylated by CK2 in vitro. Mutating these serine residues attenuates the ability of Smo to transduce high level Hh signaling activity in vivo. Furthermore, we found that CK2 plays an additional positive role downstream of Smo by regulating the stability of full-length Cubitus interruptus (Ci). CK2β RNAi promotes Ci degradation whereas coexpressing CK2α and CK2β increases the half-life of Ci. We showed that CK2 prevents Ci ubiquitination and degradation by the proteasome. Thus, CK2 promotes Hh signaling activity by regulating multiple pathway components.
Collapse
Affiliation(s)
- Hongge Jia
- Department of Molecular and Cellular Biochemistry, Markey Cancer Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Villani RM, Adolphe C, Palmer J, Waters MJ, Wainwright BJ. Patched1 Inhibits Epidermal Progenitor Cell Expansion and Basal Cell Carcinoma Formation by Limiting Igfbp2 Activity. Cancer Prev Res (Phila) 2010; 3:1222-34. [DOI: 10.1158/1940-6207.capr-10-0082] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Dessaud E, Ribes V, Balaskas N, Yang LL, Pierani A, Kicheva A, Novitch BG, Briscoe J, Sasai N. Dynamic assignment and maintenance of positional identity in the ventral neural tube by the morphogen sonic hedgehog. PLoS Biol 2010; 8:e1000382. [PMID: 20532235 PMCID: PMC2879390 DOI: 10.1371/journal.pbio.1000382] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 04/20/2010] [Indexed: 12/31/2022] Open
Abstract
Morphogens are secreted signalling molecules that act in a graded manner to control the pattern of cellular differentiation in developing tissues. An example is Sonic hedgehog (Shh), which acts in several developing vertebrate tissues, including the central nervous system, to provide positional information during embryonic patterning. Here we address how Shh signalling assigns the positional identities of distinct neuronal subtype progenitors throughout the ventral neural tube. Assays of intracellular signal transduction and gene expression indicate that the duration as well as level of signalling is critical for morphogen interpretation. Progenitors of the ventral neuronal subtypes are established sequentially, with progressively more ventral identities requiring correspondingly higher levels and longer periods of Shh signalling. Moreover, cells remain sensitive to changes in Shh signalling for an extended time, reverting to antecedent identities if signalling levels fall below a threshold. Thus, the duration of signalling is important not only for the assignment but also for the refinement and maintenance of positional identity. Together the data suggest a dynamic model for ventral neural tube patterning in which positional information corresponds to the time integral of Shh signalling. This suggests an alternative to conventional models of morphogen action that rely solely on the level of signalling.
Collapse
Affiliation(s)
- Eric Dessaud
- Developmental Neurobiology, MRC-National Institute for Medical Research, London, United Kingdom
| | - Vanessa Ribes
- Developmental Neurobiology, MRC-National Institute for Medical Research, London, United Kingdom
| | - Nikolaos Balaskas
- Developmental Neurobiology, MRC-National Institute for Medical Research, London, United Kingdom
| | - Lin Lin Yang
- Department of Neurobiology, Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Alessandra Pierani
- Institut Jacques Monod, Université Paris Diderot, Program of Development and Neurobiology, Paris, France
| | - Anna Kicheva
- Developmental Neurobiology, MRC-National Institute for Medical Research, London, United Kingdom
| | - Bennett G. Novitch
- Department of Neurobiology, Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - James Briscoe
- Developmental Neurobiology, MRC-National Institute for Medical Research, London, United Kingdom
| | - Noriaki Sasai
- Developmental Neurobiology, MRC-National Institute for Medical Research, London, United Kingdom
| |
Collapse
|
40
|
Khaliullina H, Panáková D, Eugster C, Riedel F, Carvalho M, Eaton S. Patched regulates Smoothened trafficking using lipoprotein-derived lipids. Development 2009; 136:4111-21. [PMID: 19906846 DOI: 10.1242/dev.041392] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hedgehog (Hh) is a lipoprotein-borne ligand that regulates both patterning and proliferation in a wide variety of vertebrate and invertebrate tissues. When Hh is absent, its receptor Patched (Ptc) represses Smoothened (Smo) signaling by an unknown catalytic mechanism that correlates with reduced Smo levels on the basolateral membrane. Ptc contains a sterol-sensing domain and is similar to the Niemann-Pick type C-1 protein, suggesting that Ptc might regulate lipid trafficking to repress Smo. However, no endogenous lipid regulators of Smo have yet been identified, nor has it ever been shown that Ptc actually controls lipid trafficking. Here, we show that Drosophila Ptc recruits internalized lipoproteins to Ptc-positive endosomes and that its sterol-sensing domain regulates trafficking of both lipids and Smo from this compartment. Ptc utilizes lipids derived from lipoproteins to destabilize Smo on the basolateral membrane. We propose that Ptc normally regulates Smo degradation by changing the lipid composition of endosomes through which Smo passes, and that the presence of Hh on lipoproteins inhibits utilization of their lipids by Ptc.
Collapse
Affiliation(s)
- Helena Khaliullina
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse-108, 01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Farzan SF, Stegman MA, Ogden SK, Ascano M, Black KE, Tacchelly O, Robbins DJ. A quantification of pathway components supports a novel model of Hedgehog signal transduction. J Biol Chem 2009; 284:28874-84. [PMID: 19717563 PMCID: PMC2781433 DOI: 10.1074/jbc.m109.041608] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 08/25/2009] [Indexed: 11/06/2022] Open
Abstract
The secreted protein Hedgehog (Hh) plays a critical instructional role during metazoan development. In Drosophila, Hh signaling is interpreted by a set of conserved, downstream effectors that differentially localize and interact to regulate the stability and activity of the transcription factor Cubitus interruptus. Two essential models that integrate genetic, cell biological, and biochemical information have been proposed to explain how these signaling components relate to one another within the cellular context. As the molar ratios of the signaling effectors required in each of these models are quite different, quantitating the cellular ratio of pathway components could distinguish these two models. Here, we address this important question using a set of purified protein standards to perform a quantitative analysis of Drosophila cell lysates for each downstream pathway component. We determine each component's steady-state concentration within a given cell, demonstrate the molar ratio of Hh signaling effectors differs more than two orders of magnitude and that this ratio is conserved in vivo. We find that the G-protein-coupled transmembrane protein Smoothened, an activating component, is present in limiting amounts, while a negative pathway regulator, Suppressor of Fused, is present in vast molar excess. Interestingly, despite large differences in the steady-state ratio, all downstream signaling components exist in an equimolar membrane-associated complex. We use these quantitative results to re-evaluate the current models of Hh signaling and now propose a novel model of signaling that accounts for the stoichiometric differences observed between various Hh pathway components.
Collapse
Affiliation(s)
- Shohreh F. Farzan
- From the Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755 and
| | - Melanie A. Stegman
- From the Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755 and
| | - Stacey K. Ogden
- From the Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755 and
| | - Manuel Ascano
- From the Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755 and
| | - Kendall E. Black
- From the Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755 and
| | - Ofelia Tacchelly
- From the Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755 and
| | - David J. Robbins
- From the Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755 and
- the Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire 03756
| |
Collapse
|
42
|
Baker NE, Bhattacharya A, Firth LC. Regulation of Hh signal transduction as Drosophila eye differentiation progresses. Dev Biol 2009; 335:356-66. [PMID: 19761763 DOI: 10.1016/j.ydbio.2009.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 09/01/2009] [Accepted: 09/07/2009] [Indexed: 11/16/2022]
Abstract
Differentiation of the Drosophila retina occurs as a morphogenetic furrow sweeps anteriorly across the eye imaginal disc, driven by Hedgehog secretion from photoreceptor precursors differentiating behind the furrow. A BTB protein, Roadkill, is expressed posterior to the furrow and targets the Hedgehog signal transduction component Cubitus interruptus for degradation by Cullin-3 and the proteosome. Clonal analysis and conditional mutant studies establish that roadkill transcription is activated by the EGF receptor and Ras pathway in most differentiating retinal cells, and by both EGF receptor/Ras and by Hedgehog signaling in cells that remain unspecified. These findings outline a circuit by which Hedgehog signal transduction is modified as Hedgehog signaling initiates retinal differentiation. A model is presented for regulation of the Cullin-3 and Cullin-1 pathways that modifies Hedgehog signaling as the morphogenetic furrow moves and the responses of retinal cells change.
Collapse
Affiliation(s)
- Nicholas E Baker
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | |
Collapse
|
43
|
Aanstad P, Santos N, Corbit KC, Scherz PJ, Trinh LA, Salvenmoser W, Huisken J, Reiter JF, Stainier DYR. The extracellular domain of Smoothened regulates ciliary localization and is required for high-level Hh signaling. Curr Biol 2009; 19:1034-9. [PMID: 19464178 DOI: 10.1016/j.cub.2009.04.053] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 04/10/2009] [Accepted: 04/14/2009] [Indexed: 01/01/2023]
Abstract
Members of the Hedgehog (Hh) family of secreted proteins function as morphogens to pattern developing tissues and control cell proliferation. The seven-transmembrane domain (7TM) protein Smoothened (Smo) is essential for the activation of all levels of Hh signaling. However, the mechanisms by which Smo differentially activates low- or high-level Hh signaling are not known. Here we show that a newly identified mutation in the extracellular domain (ECD) of zebrafish Smo attenuates Smo signaling. The Smo agonist purmorphamine induces the stabilization, ciliary translocation, and high-level signaling of wild-type Smo. In contrast, purmorphamine induces the stabilization but not the ciliary translocation or high-level signaling of the Smo ECD mutant protein. Surprisingly, a truncated form of Smo that lacks the cysteine-rich domain of the ECD localizes to the cilium but is unable to activate high-level Hh signaling. We also present evidence that cilia may be required for Hh signaling in early zebrafish embryos. These data indicate that the ECD, previously thought to be dispensable for vertebrate Smo function, both regulates Smo ciliary localization and is essential for high-level Hh signaling.
Collapse
Affiliation(s)
- Pia Aanstad
- Department of Biochemistry & Biophysics, Programs in Developmental Biology, Genetics, and Human Genetics, Cardiovascular Research Institute, Diabetes Center, University of California, San Francisco, CA 94158, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Norman RX, Ko HW, Huang V, Eun CM, Abler LL, Zhang Z, Sun X, Eggenschwiler JT. Tubby-like protein 3 (TULP3) regulates patterning in the mouse embryo through inhibition of Hedgehog signaling. Hum Mol Genet 2009; 18:1740-54. [PMID: 19286674 DOI: 10.1093/hmg/ddp113] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Tubby-like protein 3 (TULP3) is required for proper embryonic development in mice. Disruption of mouse Tulp3 results in morphological defects in the embryonic craniofacial regions, the spinal neural tube and the limbs. Here, we show that TULP3 functions as a novel negative regulator of Sonic hedgehog (Shh) signaling in the mouse. In Tulp3 mutants, ventral cell types in the lumbar neural tube, which acquire their identities in response to Shh signaling, are ectopically specified at the expense of dorsal cell types. Genetic epistasis experiments show that this ventralized phenotype occurs independently of Shh and the transmembrane protein Smoothened, but it is dependent on the transcription factor Gli2. The ventralized phenotype is also dependent on the kinesin II subunit Kif3A, which is required for intraflagellar transport and ciliogenesis. In addition, TULP3 is required for proper Shh-dependent limb patterning and for maintaining the correct balance between differentiation and proliferation in the neural tube. Finally, the localization of TULP3 to the tips of primary cilia raises the possibility that it regulates the Hedgehog pathway within this structure.
Collapse
Affiliation(s)
- Ryan X Norman
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Deshpande G, Godishala A, Schedl P. Ggamma1, a downstream target for the hmgcr-isoprenoid biosynthetic pathway, is required for releasing the Hedgehog ligand and directing germ cell migration. PLoS Genet 2009; 5:e1000333. [PMID: 19132091 PMCID: PMC2607556 DOI: 10.1371/journal.pgen.1000333] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Accepted: 12/09/2008] [Indexed: 11/18/2022] Open
Abstract
The isoprenoid biosynthetic pathway leading from the production of mevalonate by HMGCoA reductase (Hmgcr) to the geranylation of the G protein subunit, Ggamma1, plays an important role in cardiac development in the fly. Hmgcr has also been implicated in the release of the signaling molecule Hedgehog (Hh) from hh expressing cells and in the production of an attractant that directs primordial germ cells to migrate to the somatic gonadal precursor cells (SGPs). The studies reported here indicate that this same hmgcr-->Ggamma1 pathway provides a novel post-translational mechanism for modulating the range and activity of the Hh signal produced by hh expressing cells. We show that, like hmgcr, ggamma1 and quemao (which encodes the enzyme, geranylgeranyl diphosphate synthetase, that produces the substrate for geranylation of Ggamma1) are components of the hh signaling pathway and are required for the efficient release of the Hh ligand from hh expressing cells. We also show that the hmgcr-->Ggamma1 pathway is linked to production of the germ cell attractant by the SGPs through its ability to enhance the potency of the Hh signal. We show that germ cell migration is disrupted by the loss or gain of ggamma1 activity, by trans-heterozygous combinations between ggamma1 and either hmgcr or hh mutations, and by ectopic expression of dominant negative Ggamma1 proteins that cannot be geranylated.
Collapse
Affiliation(s)
- Girish Deshpande
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Anuradha Godishala
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Paul Schedl
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
46
|
Firth LC, Baker NE. Retinal determination genes as targets and possible effectors of extracellular signals. Dev Biol 2008; 327:366-75. [PMID: 19135045 DOI: 10.1016/j.ydbio.2008.12.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 12/15/2008] [Accepted: 12/16/2008] [Indexed: 01/01/2023]
Abstract
Retinal determination genes are sufficient to specify eyes in ectopic locations, raising the question of how these master regulatory genes define an eye developmental field. Genetic mosaic studies establish that expression of the retinal determination genes eyeless, teashirt, homothorax, eyes absent, sine oculis, and dachshund are each regulated by combinations of Dpp, Hh, N, Wg, and Ras signals in Drosophila. Dpp and Hh control eyeless, teashirt, sine oculis, and dachshund expression, Dpp and Ras control homothorax, and all the signaling pathways affect eyes absent expression. These results suggest that eye-specific development uses retinal determination gene expression to relay positional information to eye target genes, because the distinct, overlapping patterns of retinal determination gene expression reflect the activities of the extracellular signaling pathways.
Collapse
Affiliation(s)
- Lucy C Firth
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | | |
Collapse
|
47
|
Costal2 functions as a kinesin-like protein in the hedgehog signal transduction pathway. Curr Biol 2008; 18:1215-20. [PMID: 18691888 DOI: 10.1016/j.cub.2008.07.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 07/02/2008] [Accepted: 07/04/2008] [Indexed: 11/21/2022]
Abstract
The Hedgehog (Hh) signaling pathway initiates an evolutionarily conserved developmental program required for the proper patterning of many tissues [1]. Although Costal2 (Cos2) is a requisite component of the Hh pathway, its mechanistic role is not well understood. Because of its primary sequence, Cos2 was initially predicted to function as a kinesin-like protein [2]. However, evidence showing that Cos2 function might require kinesin-like properties has been lacking [2-6]. Thus, the prevailing dogma in the field is that Cos2 functions solely as a scaffolding protein [7, 8]. Here, we show that Cos2 motility is required for its biological function and that this motility may be Hh regulated. We show that Cos2 motility requires an active motor domain, ATP, and microtubules. Additionally, Cos2 recruits and transports other components of the Hh signaling pathway, including the transcription factor Cubitus interruptus (Ci). Drosophila expressing cos2 mutations that encode proteins that lack motility are attenuated in their ability to regulate Ci activity and exhibit phenotypes consistent with attenuated Cos2 function [9]. Combined, these results demonstrate that Cos2 motility plays an important role in its function, regulating the amounts and activity of Ci that ultimately interpret the level of Hh to which cells are exposed.
Collapse
|
48
|
Hosoya T, Arai MA, Koyano T, Kowithayakorn T, Ishibashi M. Naturally occurring small-molecule inhibitors of hedgehog/GLI-mediated transcription. Chembiochem 2008; 9:1082-92. [PMID: 18357592 DOI: 10.1002/cbic.200700511] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The aberrant hedgehog (Hh)/GLI signaling pathway causes the formation and progression of a variety of tumors. To search for Hh/GLI inhibitors, we screened for naturally occurring inhibitors of the transcriptional activator GLI1 by using a cell-based assay. We identified zerumbone (1), zerumbone epoxide (2), staurosporinone (9), 6-hydroxystaurosporinone (10), arcyriaflavin C (11) and 5,6-dihydroxyarcyriaflavin A (12) as inhibitors of GLI-mediated transcription. In addition, we isolated physalins F (17) and B (18) from Physalis minima, which are also potent inhibitors. These compounds also inhibited GLI2-mediated transactivation. Semiquantitative RT-PCR and Western blotting analysis further revealed that 1, 9, 17, and 18 decreased Hh-related component expressions. We also show that inhibitors of GLI-mediated transactivation reduce the level of the antiapoptosis Bcl2 expression. Finally, these identified compounds were cytotoxic to PANC1 pancreatic cancer cells, which express Hh/GLI components. These results strongly suggest that the cytotoxicity of the compounds to PANC1 cells correlates with their inhibition of GLI-mediated transcription.
Collapse
Affiliation(s)
- Takahiro Hosoya
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan
| | | | | | | | | |
Collapse
|
49
|
Schlichting K, Dahmann C. Hedgehog and Dpp signaling induce cadherin Cad86C expression in the morphogenetic furrow during Drosophila eye development. Mech Dev 2008; 125:712-28. [PMID: 18539010 DOI: 10.1016/j.mod.2008.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Revised: 03/28/2008] [Accepted: 04/19/2008] [Indexed: 01/09/2023]
Abstract
During Drosophila eye development, cell differentiation is preceded by the formation of a morphogenetic furrow, which progresses across the epithelium from posterior to anterior. Cells within the morphogenetic furrow are apically constricted and shortened along their apical-basal axis. However, how these cell shape changes and, thus, the progression of the morphogenetic furrow are controlled is not well understood. Here we show that cells simultaneously lacking Hedgehog and Dpp signal transduction fail to shorten and do not enter the morphogenetic furrow. Moreover, we have identified a gene, cadherin Cad86C, which is highly expressed in cells of the leading flank of the morphogenetic furrow. Ectopic activation of either the Hedgehog or Dpp signal transduction pathway results in elevated Cad86C expression. Conversely, simultaneous loss of both Hedgehog and Dpp signal transduction leads to decreased Cad86C expression. Finally, ectopic expression of Cad86C in either eye-antennal imaginal discs or wing imaginal discs results in apical constriction and shortening of cells. We conclude that Hedgehog and Dpp signaling promote the shortening of cells within the morphogenetic furrow. Induction of Cad86C expression might be one mechanism through which Hedgehog and Dpp promote these cell shape changes.
Collapse
Affiliation(s)
- Karin Schlichting
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | | |
Collapse
|
50
|
Lagerström MC, Schiöth HB. Structural diversity of G protein-coupled receptors and significance for drug discovery. Nat Rev Drug Discov 2008; 7:339-57. [PMID: 18382464 DOI: 10.1038/nrd2518] [Citation(s) in RCA: 1063] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of membrane-bound receptors and also the targets of many drugs. Understanding of the functional significance of the wide structural diversity of GPCRs has been aided considerably in recent years by the sequencing of the human genome and by structural studies, and has important implications for the future therapeutic potential of targeting this receptor family. This article aims to provide a comprehensive overview of the five main human GPCR families--Rhodopsin, Secretin, Adhesion, Glutamate and Frizzled/Taste2--with a focus on gene repertoire, general ligand preference, common and unique structural features, and the potential for future drug discovery.
Collapse
Affiliation(s)
- Malin C Lagerström
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, BOX 593, 751 24, Uppsala, Sweden
| | | |
Collapse
|