551
|
Isolation and characterization of a secreted, cell-surface glycoprotein SCUBE2 from humans. Biochem J 2009; 422:119-28. [DOI: 10.1042/bj20090341] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
SCUBE2 [signal peptide, CUB domain, EGF (epidermal growth factor)-like protein 2] belongs to an evolutionarily conserved SCUBE protein family, which possesses domain organization characteristic of an N-terminal signal peptide sequence followed by nine EGF-like repeats, a spacer region, three cysteine-rich repeat motifs, and one CUB domain at the C-terminus. Despite several genetic analyses suggesting that the zebrafish orthologue of the mammalian SCUBE2 gene participates in HH (Hedgehog) signalling, the complete full-length cDNA and biochemical function for mammalian SCUBE2 on HH signalling remains uninvestigated. In the present study, we isolated the full-length cDNA and studied the role of human SCUBE2 in the HH signalling cascade. When overexpressed, recombinant human SCUBE2 manifests as a secreted surface-anchored glycoprotein. Deletion mapping analysis defines the critical role of the spacer region and/or cysteine-rich repeats for membrane association. Further biochemical analyses and functional reporter assays demonstrated that human SCUBE2 can specifically interact with SHH (Sonic Hedgehog) and SHH receptor PTCH1 (Patched-1), and enhance the SHH signalling activity within the cholesterol-rich raft microdomains of the plasma membranes. Together, our results reveal that human SCUBE2 is a novel positive component of the HH signal, acting upstream of ligand binding at the plasma membrane. Thus human SCUBE2 could play important roles in HH-related biology and pathology, such as during organ development and tumour progression.
Collapse
|
552
|
Kim WK, Meliton V, Park KW, Hong C, Tontonoz P, Niewiadomski P, Waschek JA, Tetradis S, Parhami F. Negative regulation of Hedgehog signaling by liver X receptors. Mol Endocrinol 2009; 23:1532-43. [PMID: 19608643 DOI: 10.1210/me.2008-0453] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Hedgehog (Hh) signaling is indispensable in embryonic development, and its dysregulated activity results in severe developmental disorders as shown by genetic models of naturally occurring mutations in animal and human pathologies. Hh signaling also functions in postembryonic development and adult tissue homeostasis, and its aberrant activity causes various human cancers. Better understanding of molecular regulators of Hh signaling is of fundamental importance in finding new strategies for pathway modulation. Here, we identify liver X receptors (LXRs), members of the nuclear hormone receptor family, as previously unrecognized negative regulators of Hh signaling. Activation of LXR by specific pharmacological ligands, TO901317 and GW3965, inhibited the responses of pluripotent bone marrow stromal cells and calvaria organ cultures to sonic Hh, resulting in the inhibition of expression of Hh-target genes, Gli1 and Patched1, and Gli-dependent transcriptional activity. Moreover, LXR ligands inhibited sonic Hh-induced differentiation of bone marrow stromal cells into osteoblasts. Elimination of LXRs by small interfering RNA inhibited ligand-induced inhibition of Hh target gene expression. Furthermore, LXR ligand did not inhibit Hh responsiveness in mouse embryonic fibroblasts that do not express LXRs, whereas introduction of LXR into these cells reestablished the inhibitory effects. Daily oral administration of TO901317 to mice after 3 d significantly inhibited baseline Hh target-gene expression in liver, lung, and spleen. Given the importance of modulating Hh signaling in various physiological and pathological settings, our findings suggest that pharmacological targeting of LXRs may be a novel strategy for Hh pathway modulation.
Collapse
Affiliation(s)
- Woo-Kyun Kim
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
553
|
Medina V, Calvo MB, Díaz-Prado S, Espada J. Hedgehog signalling as a target in cancer stem cells. Clin Transl Oncol 2009; 11:199-207. [PMID: 19380296 DOI: 10.1007/s12094-009-0341-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hedgehog (Hh) is one of the most important signalling pathways. Together with the Wnt, TGF-Beta/BMP and Notch pathways, it is involved in both embryonic development and adult tissue homeostasis. This is because Hh plays a central role in the proliferative control and differentiation of both embryonic stem cells and adult stem cells. In this way, an alteration in the Hh pathway, either by misexpression of components of that pathway or by changes in the expression of other cellular components that interfere with the Hh signalling system, may trigger the development of several types of cancer. This occurs because normal stem cells or their intermediaries toward differentiated mature cells are not part of the normal proliferative/ differentiation balance and begin to expand without control, triggering the generation of the so-called cancer stem cells. In this review, we will focus on the molecular aspects and the role of Hh signalling in normal tissues and in tumour development.
Collapse
Affiliation(s)
- Vanessa Medina
- Oncology Research Unit, University Hospital A Coruña, A Coruña, Spain.
| | | | | | | |
Collapse
|
554
|
Affiliation(s)
- Neeraj Mahindroo
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA.
| | | | | |
Collapse
|
555
|
Mizuarai S, Kawagishi A, Kotani H. Inhibition of p70S6K2 down-regulates Hedgehog/GLI pathway in non-small cell lung cancer cell lines. Mol Cancer 2009; 8:44. [PMID: 19575820 PMCID: PMC2714036 DOI: 10.1186/1476-4598-8-44] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2009] [Accepted: 07/06/2009] [Indexed: 12/21/2022] Open
Abstract
Background The Hedgehog (HH) pathway promotes tumorigenesis in a diversity of cancers. Activation of the HH signaling pathway is caused by overexpression of HH ligands or mutations in the components of the HH/GLI1 cascade, which lead to increased transactivation of GLI transcription factors. Although negative kinase regulators that antagonize the activity of GLI transcription factors have been reported, including GSK3β, PKA and CK1s, little is known regarding positive kinase regulators that are suitable for use on cancer therapeutic targets. The present study attempted to identify kinases whose silencing inhibits HH/GLI signalling in non-small cell lung cancer (NSCLC). Results To find positive kinase regulators in the HH pathway, kinome-wide siRNA screening was performed in a NSCLC cell line, A549, harboring the GLI regulatory reporter gene. This showed that p70S6K2-silencing remarkably reduced GLI reporter gene activity. The decrease in the activity of the HH pathway caused by p70S6K2-inhibition was accompanied by significant reduction in cell viability. We next investigated the mechanism for p70S6K2-mediated inhibition of GLI1 transcription by hypothesizing that GSK3β, a negative regulator of the HH pathway, is activated upon p70S6K2-silencing. We found that phosphorylated-GSK3β (Ser9) was reduced by p70S6K2-silencing, causing a decreased level of GLI1 protein. Finally, to further confirm the involvement of p70S6K2 in GLI1 signaling, down-regulation in GLI-mediated transcription by PI3KCA-inhibition was confirmed, establishing the pivotal role of the PI3K/p70S6K2 pathway in GLI1 cascade regulation. Conclusion We report herein that inhibition of p70S6K2, known as a downstream effector of the PI3K pathway, remarkably decreases GLI-mediated transactivation in NSCLC by reducing phosphorylated-GSK3β followed by GLI1 degradation. These results infer that p70S6K2 is a potential therapeutic target for NSCLC with hyperactivated HH/GLI pathway.
Collapse
Affiliation(s)
- Shinji Mizuarai
- Department of Oncology, Tsukuba Research Institute, Merck Research Laboratories, Banyu Pharmaceutical Co Ltd, Tsukuba, Ibaraki, Japan.
| | | | | |
Collapse
|
556
|
Alman BA. Multiple hereditary exostosis and hedgehog signaling: implications for novel therapies. J Bone Joint Surg Am 2009; 91 Suppl 4:63-7. [PMID: 19571070 DOI: 10.2106/jbjs.i.00301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Benjamin A Alman
- Division of Orthopaedic Surgery, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
557
|
Chakrabarti D. A methods-based biotechnology course for undergraduates. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2009; 37:227-231. [PMID: 21567741 DOI: 10.1002/bmb.20302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
This new course in biotechnology for upper division undergraduates provides a comprehensive overview of the process of drug discovery that is relevant to biopharmaceutical industry. The laboratory exercises train students in both cell-free and cell-based assays. Oral presentations by the students delve into recent progress in drug discovery. Combination of lectures, hands-on experiments, oral presentations, and accurate recording of laboratory data provides students a thorough training in biotechnology that better prepares them for the job market.
Collapse
Affiliation(s)
- Debopam Chakrabarti
- Department of Molecular Biology and Microbiology, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida 32826.
| |
Collapse
|
558
|
Büttner A, Seifert K, Cottin T, Sarli V, Tzagkaroulaki L, Scholz S, Giannis A. Synthesis and biological evaluation of SANT-2 and analogues as inhibitors of the hedgehog signaling pathway. Bioorg Med Chem 2009; 17:4943-54. [PMID: 19541490 DOI: 10.1016/j.bmc.2009.06.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 05/29/2009] [Accepted: 06/02/2009] [Indexed: 12/21/2022]
Abstract
Hedgehog (Hh) signaling plays an important role in cell signaling of embryonic development and adult tissue homeostasis. In vertebrates, the hh gene encodes three different unique proteins: sonic hedgehog (Shh), desert hedgehog (Dhh) and indian hedgehog (Ihh). Disruption of the Hh signaling pathway leads to severe disorders in the development of vertebrates whereas aberrant activation of the Hh pathway has been associated with several malignancies including Gorlin syndrome (a disorder predisposing to basal cell carcinoma, medulloblastoma and rhabdomyosarcoma), prostate, pancreatic and breast cancers. In vivo evidence suggests the antagonism of excessive Hh signaling provides a route to unique mechanism-based anti-cancer therapies. Recently the small molecule SANT-2 was identified as a potent antagonist of Hh-signaling pathway. Here, we describe the synthesis, SAR studies as well as biological evaluation of SANT-2 and its analogues. Fifteen SANT-2 derivatives were synthesized and analyzed for their interference with the expression of the Hh target gene Gli1 in a reporter gene assay. By comparison of structure and activity important molecular descriptors for Gli inhibition could be identified. Furthermore we identified derivative TC-132 that was slightly more potent than the parent compound SANT-2. Selected compounds were tested for Hh related teratogenic effects in the small teleost model medaka. Albeit Gli expression has indicated a 16-fold higher Hh-inhibiting activity than observed for the plant alkaloid cyclopamine, none of the tested compounds were able to induce the cyclopamine-specific phenotype in the medaka assay.
Collapse
Affiliation(s)
- Anita Büttner
- University of Leipzig, Institute for Organic Chemistry, Johannisallee 29, D-04103 Leipzig, Germany
| | | | | | | | | | | | | |
Collapse
|
559
|
Hofmann I, Stover EH, Cullen DE, Mao J, Morgan KJ, Lee BH, Kharas MG, Miller PG, Cornejo MG, Okabe R, Armstrong SA, Ghilardi N, Gould S, de Sauvage FJ, McMahon AP, Gilliland DG. Hedgehog signaling is dispensable for adult murine hematopoietic stem cell function and hematopoiesis. Cell Stem Cell 2009; 4:559-67. [PMID: 19497284 PMCID: PMC3065323 DOI: 10.1016/j.stem.2009.03.016] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 03/04/2009] [Accepted: 03/26/2009] [Indexed: 12/21/2022]
Abstract
We report the unexpected finding that loss of Hh signaling through conditional deletion of Smoothened (Smo) in the adult hematopoietic compartment has no apparent effect on adult hematopoiesis, including peripheral blood count, number or cell-cycle status of stem or progenitor cells, hematopoietic colony-forming potential, long-term repopulating activity in competitive repopulation assays, or stress response to serial 5-fluorouracil treatment. Furthermore, pharmacologic inhibition of Hh signaling with a potent and selective small molecule antagonist has no substantive effect on hematopoiesis in the mouse. In addition, Hh signaling is not required for the development of MLL-AF9-mediated acute myeloid leukemia (AML). Taken together, these data demonstrate that Hh signaling is dispensable for normal hematopoietic development and hematopoietic stem cell function, indicating that targeting of Hh signaling in solid tumors is not likely to result in hematopoietic toxicity. Furthermore, the Hh pathway may not be a compelling target in certain hematopoietic malignancies.
Collapse
Affiliation(s)
- Inga Hofmann
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Children’s Hospital Boston, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Elizabeth H. Stover
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Dana E. Cullen
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Junhao Mao
- Department of Molecular and Cellular Biology, 16 Divinity Avenue, Harvard University, Cambridge, MA 02138, USA
| | - Kelly J. Morgan
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Benjamin H. Lee
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Michael G. Kharas
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Peter G. Miller
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Melanie G. Cornejo
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Rachel Okabe
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Scott A. Armstrong
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Children’s Hospital Boston, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Nico Ghilardi
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stephen Gould
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Andrew P. McMahon
- Department of Molecular and Cellular Biology, 16 Divinity Avenue, Harvard University, Cambridge, MA 02138, USA
| | - D. Gary Gilliland
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Harvard Stem Cell Institute, 42 Church Street, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD 20815-6789, USA
| |
Collapse
|
560
|
Abstract
A diverse set of cellular defects, presumably elicited by multiple genetic alterations, underlies cancer development. Aberrant Hedgehog (Hh) signaling has recently been implicated in the development and maintenance of breast cancer. However, evidence conclusively showing that activated Hh signaling can induce mammary tumors is lacking. We now show that transgenic expression of the Hh effector protein GLI1 under the regulation of the mouse mammary tumor virus promoter, expressed in the mouse mammary gland, is associated with the appearance of hyperplastic lesions, defective terminal end buds, and tumor development. The GLI1-induced tumors are histologically heterogeneous and involve the expansion of a population of epithelial cells expressing the progenitor cell markers keratin 6 and Bmi-1. Moreover, tumor cells express genes involved in proliferation, cell survival, and metastasis. GLI1-induced tumors do not fully regress following transgene deinduction, indicating that some tumors develop and are maintained autonomously, independent of sustained transgenic GLI1 expression. The data strongly support a role of Hh/GLI signaling in breast cancer development and suggest that inhibition of this signaling pathway represents a new therapeutic opportunity for limiting tumorigenesis and early tumorigenic progression.
Collapse
Affiliation(s)
- Marie Fiaschi
- Center for Biosciences, Department of Biosciences, Karolinska Institutet,Novum, SE-141 57 Huddinge, Sweden
| | | | | | | |
Collapse
|
561
|
Barginear MF, Leung M, Budman DR. The hedgehog pathway as a therapeutic target for treatment of breast cancer. Breast Cancer Res Treat 2009; 116:239-46. [PMID: 19479372 DOI: 10.1007/s10549-009-0423-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 05/11/2009] [Indexed: 12/18/2022]
Abstract
The Hedgehog (Hh) signaling pathway plays a key role in a variety of processes, such as embryogenesis and maintenance of adult tissue homeostasis. It is also becoming increasingly clear that this pathway can have a crucial role in tumorigenesis. Most recently, the Hh signaling pathway has been implicated in the development and maintenance of breast cancer. Here we review Hh signaling, advances in small molecule and antibody-based inhibitors targeting the Hh pathway, and dysregulation of the Hh signaling pathway in breast cancer.
Collapse
Affiliation(s)
- M F Barginear
- The Breast Cancer Medicine Program at the Tisch Cancer Institute, The Mount Sinai School of Medicine, New York, NY, USA.
| | | | | |
Collapse
|
562
|
Scales SJ, de Sauvage FJ. Mechanisms of Hedgehog pathway activation in cancer and implications for therapy. Trends Pharmacol Sci 2009; 30:303-12. [PMID: 19443052 DOI: 10.1016/j.tips.2009.03.007] [Citation(s) in RCA: 521] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 03/24/2009] [Accepted: 03/25/2009] [Indexed: 02/08/2023]
Abstract
The Hedgehog (Hh) signaling pathway regulates body patterning and organ development during embryogenesis. In adults the Hh pathway is mainly quiescent, with the exception of roles in tissue maintenance and repair, and its inappropriate reactivation has been linked to several disparate human cancers. In addition to cancers with mutations in components of the Hh pathway, Hh ligand-dependent cancers have been proposed to respond to Hh in an autocrine manner. More recent findings that Hh might instead signal in a paracrine manner from the tumor to the surrounding stroma or in cancer stem cells alter our understanding of Hh mechanisms in cancer, with important implications for choice of preclinical tumor models, drug screening, patient selection and therapeutic intervention. We review here the roles of the Hh pathway in cancer, Hh pathway inhibitors (HPIs) and early clinical trial results using a novel small molecule HPI, GDC-0449.
Collapse
Affiliation(s)
- Suzie J Scales
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | |
Collapse
|
563
|
Feldmann G, Rauenzahn S, Maitra A. In vitro models of pancreatic cancer for translational oncology research. Expert Opin Drug Discov 2009; 4:429-443. [PMID: 20160967 PMCID: PMC2760831 DOI: 10.1517/17460440902821657] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND: Pancreatic cancer is a disease of near uniform fatality and the overwhelming majority of patients succumb to their advanced malignancy within a few months of diagnosis. Despite considerable advances in our understanding of molecular mechanisms underlying pancreatic carcinogenesis, this knowledge has not yet been fully translated into clinically available treatment strategies that yield significant improvements in disease free or overall survival. OBJECTIVE: Cell line-based in vitro model systems provide powerful tools to identify potential molecular targets for therapeutic intervention as well as for initial pre-clinical evaluation of novel drug candidates. Here we provide a brief overview of recent literature on cell line-based model systems of pancreatic cancer and their application in the search for novel therapeutics against this vicious disease. CONCLUSION: While in vitro models of pancreatic cancer are of tremendous value for genetic studies and initial functional screenings in drug discovery, they carry several imanent drawbacks and are often poor in predicting therapeutic response in humans. Therefore, in most instances they are successfully exploited to generate hypothesis and identify molecular targets for novel therapeutics, which are subsequently subject to further in-depth characterization using more advanced in vivo model systems and clinical trials.
Collapse
Affiliation(s)
- Georg Feldmann
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
564
|
Recent insights into the molecular mechanisms involved in aging and the malignant transformation of adult stem/progenitor cells and their therapeutic implications. Ageing Res Rev 2009; 8:94-112. [PMID: 19114129 DOI: 10.1016/j.arr.2008.12.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 12/04/2008] [Indexed: 02/07/2023]
Abstract
Recent advancements in tissue-resident adult stem/progenitor cell research have revealed that enhanced telomere attrition, oxidative stress, ultraviolet radiation exposure and oncogenic events leading to severe DNA damages and genomic instability may occur in these immature and regenerative cells during chronological aging. Particularly, the alterations in key signaling components controlling their self-renewal capacity and an up-regulation of tumor suppressor gene products such as p16(INK4A), p19(ARF), ataxia-telangiectasia mutated (ATM) kinase, p53 and/or the forkhead box O (FOXOs) family of transcription factors may result in their dysfunctions, growth arrest and senescence or apoptotic death during the aging process. These molecular events may culminate in a progressive decline in the regenerative functions and the number of tissue-resident adult stem/progenitor cells, and age-related disease development. Conversely, the telomerase re-activation and accumulation of numerous genetic and/or epigenetic alterations in adult stem/progenitor cells with advancing age may result in their immortalization and malignant transformation into highly leukemic or tumorigenic cancer-initiating cells and cancer initiation. Therefore, the cell-replacement and gene therapies and molecular targeting of aged and dysfunctional adult stem/progenitor cells including their malignant counterpart, cancer-initiating cells, hold great promise for treating and even curing diverse devastating human diseases. These diseases include premature aging diseases, hematopoietic, cardiovascular, musculoskeletal, pulmonary, ocular, urogenital, neurodegenerative and skin disorders and aggressive and recurrent cancers.
Collapse
|
565
|
Lichtenauer UD, Beuschlein F. The tumor stem cell concept-implications for endocrine tumors? Mol Cell Endocrinol 2009; 300:158-63. [PMID: 19027822 DOI: 10.1016/j.mce.2008.10.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 10/19/2008] [Accepted: 10/22/2008] [Indexed: 12/21/2022]
Abstract
The cancer stem cell hypothesis has recently evolved from an increasing body of evidence suggesting that in some cancers a small population of tumor cells with stem cell-like properties represents a critical component that dictates the malignant behavior of a given tumor. These observations challenge classical cancer biology and its theory, that tumor growth is mainly based on genomic alterations followed by modulation of cell cycle pathways, which finally result in uncontrolled clonal proliferation. Over the last few years, much progress in the field of tumor stem cells has been achieved in non-endocrine malignancies. In this review, we summarize the existing evidence regarding the tumor stem cell concept for tumor pathophysiology in general and highlight current models that have the potential to further impact research on endocrine tumors.
Collapse
Affiliation(s)
- Urs D Lichtenauer
- Medical Clinic, University Hospital Innenstadt, Ludwig Maximilians University, Munich, Germany
| | | |
Collapse
|
566
|
Rohatgi R, Milenkovic L, Corcoran RB, Scott MP. Hedgehog signal transduction by Smoothened: pharmacologic evidence for a 2-step activation process. Proc Natl Acad Sci U S A 2009; 106:3196-201. [PMID: 19218434 PMCID: PMC2642660 DOI: 10.1073/pnas.0813373106] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Indexed: 11/18/2022] Open
Abstract
The Hedgehog (Hh) signaling pathway controls growth, cell fate decisions, and morphogenesis during development. Damage to Hh transduction machinery can lead to birth defects and cancer. The transmembrane protein Smoothened (Smo) relays the Hh signal and is an important drug target in cancer. Smo enrichment in primary cilia is thought to drive activation of target genes. Using small-molecule agonists and antagonists to dissect Smo function, we find that Smo enrichment in cilia is not sufficient for signaling and a distinct second step is required for full activation. This 2-step mechanism--localization followed by activation--has direct implications for the design and use of anticancer therapeutics targeted against Smo.
Collapse
Affiliation(s)
- Rajat Rohatgi
- Departments of Developmental Biology
- Genetics
- Bioengineering, and
- Oncology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| | | | | | | |
Collapse
|
567
|
Gudjonsson JE, Aphale A, Grachtchouk M, Ding J, Nair RP, Wang T, Voorhees JJ, Dlugosz AA, Elder JT. Lack of evidence for activation of the hedgehog pathway in psoriasis. J Invest Dermatol 2009; 129:635-40. [PMID: 18754037 PMCID: PMC2771222 DOI: 10.1038/jid.2008.266] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent reports have suggested that the hedgehog (Hh) pathway is activated in lesional psoriatic skin, and that treatment with the Hh pathway antagonist cyclopamine may lead to rapid resolution of the disease. To assess Hh pathway activity in psoriasis, we isolated RNA from lesional and uninvolved skin of 58 psoriatic patients, and from 63 normal control subjects, and subjected these samples to global gene expression profiling on Affymetrix HU133 Plus 2.0 gene arrays. We were especially interested in Hh target genes (PTCH1 and GLI1), whose expression is elevated in response to Hh signaling. The microarray data demonstrated downregulation of PTCH1 expression in uninvolved and lesional skin (1.1-fold and 2-fold, respectively; P<0.0001). Additionally GLI1 mRNA was downregulated in lesional skin (1.7 fold; P<0.05). No significant changes were observed between lesional and uninvolved skin for the Hh ligands or Smoothened. Quantitative PCR confirmed these findings. In situ hybridization for GLI1 and PTCH1 was positive in basal cell carcinoma tumor cells, but was negligible in uninvolved or lesional psoriatic skin. The absence of elevated Hh target gene expression in lesional psoriatic skin indicates that the Hh pathway is not activated in this disease, raising questions regarding the proposed use of Hh antagonists as antipsoriatic agents.
Collapse
Affiliation(s)
- Johann E Gudjonsson
- Department of Dermatology, University of Michigan Medical Center, Ann Arbor, Michigan, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
568
|
Stanton BZ, Peng LF, Maloof N, Nakai K, Wang X, Herlihy KM, Duffner JL, Taveras KM, Hyman JM, Lee SW, Koehler AN, Chen JK, Fox JL, Mandinova A, Schreiber SL. A small molecule that binds Hedgehog and blocks its signaling in human cells. Nat Chem Biol 2009; 5:154-6. [PMID: 19151731 PMCID: PMC2770933 DOI: 10.1038/nchembio.142] [Citation(s) in RCA: 238] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 12/30/2008] [Indexed: 12/13/2022]
Abstract
Small-molecule inhibition of extracellular proteins that activate membrane receptors has proven to be extremely challenging. Diversity-oriented synthesis and small-molecule microarrays enabled the discovery of robotnikinin, a small molecule that binds the extracellular Sonic hedgehog (Shh) protein and blocks Shh signaling in cell lines, human primary keratinocytes and a synthetic model of human skin. Shh pathway activity is rescued by small-molecule agonists of Smoothened, which functions immediately downstream of the Shh receptor Patched.
Collapse
Affiliation(s)
- Benjamin Z. Stanton
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts 02142
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138
| | - Lee F. Peng
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts 02142
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138
- GI Unit, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114
| | - Nicole Maloof
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts 02142
| | - Kazuo Nakai
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138
| | - Xiang Wang
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts 02142
| | - Kara M. Herlihy
- Biacore, Inc., 200 Centennial Ave., Suite 100, Piscataway, NJ 08854
| | - Jay L. Duffner
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts 02142
| | - Kennedy M. Taveras
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts 02142
| | - Joel M. Hyman
- Department of Chemical and Systems Biology, Stanford University School of Medicine, 269 Campus Drive, CCSR 3155, Stanford, California, 94305
| | - Sam W. Lee
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Angela N. Koehler
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts 02142
| | - James K. Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, 269 Campus Drive, CCSR 3155, Stanford, California, 94305
| | - Julia L. Fox
- Department of Stem Cell and Regenerative Biology, Harvard University, 42 Church St, Cambridge, MA 02138
| | - Anna Mandinova
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Stuart L. Schreiber
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts 02142
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138
- The Howard Hughes Medical Institute
| |
Collapse
|
569
|
Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc Natl Acad Sci U S A 2009; 106:4254-9. [PMID: 19246386 DOI: 10.1073/pnas.0813203106] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Hedgehog (Hh) pathway has been implicated in pancreatic cancer but its role remains controversial. To delineate the cell populations able to respond to Hh ligand stimulation, we expressed an oncogenic allele of Smoothened (SmoM2) to cell autonomously activate Hh signaling in the mouse pancreas. Surprisingly, we found that expression of SmoM2 in epithelial cells was not able to activate the pathway and had no impact on pancreatic development or neoplasia. In contrast, activation of Smo in the mesenchyme led to Hh pathway activation, indicating that only the tumor stroma is competent to transduce the Hh signal. Using a Ptc-LacZ reporter mouse, we show that Hh signaling is active in stromal cells surrounding Hh-expressing tumor epithelium in various mouse pancreatic cancer models. Activation of the Hh pathway in the tumor stroma of human pancreatic and metastatic cancer specimens was confirmed by quantitative RT-PCR of microdissected tissue samples. These data support a paracrine model of Hh-mediated tumorigenesis, in which tumor cells secrete Hh ligand to induce tumor-promoting Hh target genes in adjacent stroma.
Collapse
|
570
|
Kasper M, Jaks V, Fiaschi M, Toftgård R. Hedgehog signalling in breast cancer. Carcinogenesis 2009; 30:903-11. [PMID: 19237605 DOI: 10.1093/carcin/bgp048] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common cause of cancer death among women worldwide. In order to improve the treatment of this disease, a more complete understanding of its biological basis is necessary. Since the Hedgehog (Hh) pathway was recently found to be required for growth and propagation of a number of different cancers, we discuss here the possible involvement of this pathway in the normal biology and development of cancer in the mammary gland. The use of mouse mammary cancer models has assisted the process of dissecting the mechanisms behind Hh-driven mammary tumour formation and growth. Based on recent studies, we conclude that the inhibition of Hh signalling in breast tumours may interfere with the maintenance of a putative cancer stem cell compartment and the abnormal stimulation of tumour stroma. Therefore, the components of the Hh signalling cascade may provide a set of drug targets, which could be implemented into novel combinatorial strategies for the treatment of breast cancer.
Collapse
Affiliation(s)
- Maria Kasper
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden
| | | | | | | |
Collapse
|
571
|
Laner-Plamberger S, Kaser A, Paulischta M, Hauser-Kronberger C, Eichberger T, Frischauf AM. Cooperation between GLI and JUN enhances transcription of JUN and selected GLI target genes. Oncogene 2009; 28:1639-51. [PMID: 19219074 DOI: 10.1038/onc.2009.10] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sustained Hedgehog (HH) signaling is implicated in basal cell carcinoma of the skin and other types of cancer. Here we show that GLI1 and GLI2, the main transcriptional activators of the HH pathway, directly regulate expression of the activator protein 1 (AP-1) family member JUN, a transcription factor controlling keratinocyte proliferation and skin homeostasis. Activation of the JUN promoter by GLI is dependent on a GLI-binding site and the AP-1 sites known to be involved in self-activation of JUN. Transcription of JUN is greatly enhanced in the presence of GLI and requires activated JUN protein. GLI2act is a more potent activator than GLI1 in these experiments and physical interaction with phosphorylated JUN was only detected for GLI2act. The synergistic effect of GLI and JUN extends to the activation of further GLI target genes as shown by shRNA-mediated knockdown of JUN in human keratinocytes. Some of these cooperatively activated genes are involved in cell-cycle progression, which is consistent with a significant reduction of the proliferative potential of GLI in the absence of JUN. These results suggest a novel connection between HH/GLI pathway activity and JUN, which may contribute to the oncogenic activity of HH/GLI signaling in skin.
Collapse
Affiliation(s)
- S Laner-Plamberger
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | | | | | | | | | | |
Collapse
|
572
|
Schnidar H, Eberl M, Klingler S, Mangelberger D, Kasper M, Hauser-Kronberger C, Regl G, Kroismayr R, Moriggl R, Sibilia M, Aberger F. Epidermal growth factor receptor signaling synergizes with Hedgehog/GLI in oncogenic transformation via activation of the MEK/ERK/JUN pathway. Cancer Res 2009; 69:1284-92. [PMID: 19190345 PMCID: PMC3035872 DOI: 10.1158/0008-5472.can-08-2331] [Citation(s) in RCA: 168] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Persistent activation of the Hedgehog (HH)/GLI signaling pathway has been implicated in the development of a number of human cancers. The GLI zinc finger transcription factors act at the end of the HH signaling cascade to control gene expression, and recent studies have shown that the activity of GLI proteins can be additionally modified by integration of distinct signals, such as the MEK/extracellular signal-regulated kinase (ERK) and phosphinositide-3 kinase (PI3K)/AKT pathway. However, little is known about the identity of the upstream activators of these HH/GLI interacting signaling pathways in cancer. Here, we provide evidence that integration of the HH/GLI and epidermal growth factor receptor (EGFR) pathway synergistically induces oncogenic transformation, which depends on EGFR-mediated activation of the RAS/RAF/MEK/ERK but not of the PI3K/AKT pathway. EGFR/MEK/ERK signaling induces JUN/activator protein 1 activation, which is essential for oncogenic transformation, in combination with the GLI activator forms GLI1 and GLI2. Furthermore, pharmacologic inhibition of EGFR and HH/GLI efficiently reduces growth of basal cell carcinoma (BCC) cell lines derived from mice with activated HH/GLI signaling. The results identify the synergistic integration of GLI activator function and EGFR signaling as a critical step in oncogenic transformation and provide a molecular basis for therapeutic opportunities relying on combined inhibition of the HH/GLI and EGFR/MEK/ERK/JUN pathway in BCC.
Collapse
Affiliation(s)
- Harald Schnidar
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Markus Eberl
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Stefan Klingler
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Doris Mangelberger
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Maria Kasper
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | | | - Gerhard Regl
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Renate Kroismayr
- Institute of Cancer Research, Medical University Vienna, Vienna, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Medical University Vienna, Vienna, Austria
| | - Fritz Aberger
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| |
Collapse
|
573
|
Ren Y, Cowan RG, Harman RM, Quirk SM. Dominant activation of the hedgehog signaling pathway in the ovary alters theca development and prevents ovulation. Mol Endocrinol 2009; 23:711-23. [PMID: 19196835 DOI: 10.1210/me.2008-0391] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The role of the hedgehog (HH) signaling pathway in ovarian function was examined in transgenic mice in which expression of a dominant active allele of the signal transducer smoothened (SmoM2) was directed to the ovary and Müllerian duct by cre-mediated recombination (Amhr2(cre/+)SmoM2). Mutant mice were infertile and had ovarian and reproductive tract defects. Ovaries contained follicles of all sizes and corpora lutea (CL), but oocytes were rarely recovered from the oviducts of superovulated mice and remained trapped in preovulatory follicles. Measures of luteinization did not differ. Cumulus expansion appeared disorganized, and in vitro analyses confirmed a reduced expansion index. Microarray analysis indicated that expression levels of genes typical of smooth muscle were reduced in mutant mice, and RT-PCR showed that levels of expression of muscle genes were reduced in the nongranulosa, theca-interstitial cell-enriched fraction. Whereas a layer of cells in the outer theca was positively stained for smooth muscle actin in control ovaries, this staining was reduced or absent in mutant ovaries. Expression of a number of genes in granulosa cells that are known to be important for ovulation did not differ in mutants and controls. Expression of components of the HH pathway was observed in both granulosa cells and in the nongranulosa, residual ovarian tissue and changed in response to treatment with equine chorionic gonadotropin/human gonadotropin. The results show that appropriate signaling through the HH pathway is required for development of muscle cells within the theca and that impaired muscle development is associated with failure to release the oocyte at ovulation.
Collapse
Affiliation(s)
- Yi Ren
- Department of Animal Science, Cornell University, Ithaca, NY 14853, USA
| | | | | | | |
Collapse
|
574
|
Selective translocation of intracellular Smoothened to the primary cilium in response to Hedgehog pathway modulation. Proc Natl Acad Sci U S A 2009; 106:2623-8. [PMID: 19196978 DOI: 10.1073/pnas.0812110106] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Smoothened (Smo), a 7-pass transmembrane protein, is essential for transduction of a Hedgehog (Hh) signal across the cell membrane. Smo is also the principle therapeutic target for several candidate drugs in the treatment of Hh-related diseases. Mammalian Smo translocates to the primary cilium in response to Sonic hedgehog (Shh) ligand-mediated signaling. A mechanistic understanding of Smo translocation and its interactions with drug candidates is pivotal to our understanding of Hh signaling and the design, development and application of successful drugs. We established a system in which Smo was dual-labeled with GFP and a 12-aa tag whose recognition by an enzymatic process enables the posttranslational labeling of Smo in the cell membrane within the living cell. These tools enable the simultaneous visualization of all cellular Smo, and more specifically, the cell membrane restricted subpopulation. Using this system, we demonstrate that cyclopamine, a widely used Hh antagonist, induces a cilial translocation of Smo similar to that reported for Shh ligand and several Hh agonists. In contrast, other antagonists abrogate the Shh-induced, cilial translocation of Smo. We present evidence that the majority of cilial-localized Smo originates from an intracellular source and may traffic to the primary cilium through an intraflagellar transport (IFT) pathway.
Collapse
|
575
|
El-Zaatari M, Saqui-Salces M, Waghray M, Todisco A, Merchant JL. Sonic hedgehog in gastric physiology and neoplastic transformation: friend or foe? Curr Opin Endocrinol Diabetes Obes 2009; 16:60-5. [PMID: 19104239 PMCID: PMC2895804 DOI: 10.1097/med.0b013e328320a821] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW To understand the role of sonic hedgehog (Shh) in normal gastric physiology and neoplastic transformation. RECENT FINDINGS Emerging evidence shows that gastric epithelial cells produce Shh ligand, which subsequently targets the mesenchyme. This paracrine signaling event is recapitulated by Shh-producing tumors that signal to the supporting stroma to encourage growth. Primary cilia contain components of the hedgehog signaling apparatus, and thus are typically found on responding stromal cells. SUMMARY In the stomach, Shh is produced in epithelial cells and received by responding cells in the mesenchyme. In vitro, Shh enhances gastric acid secretion and induces mucin expression. It remains to be determined whether the canonical signaling pathway mediates the observed epithelial effects. Shh expression and signaling is reduced in chronic gastritis, and Shh(-/-) embryos exhibit hyperplasia and metaplastic changes in the gastric mucosa. After its loss in the corpus, Shh is re-expressed in some gastric carcinomas typically arising in the distal stomach or antrum, suggesting that it promotes tumor growth.
Collapse
Affiliation(s)
| | | | - Megna Waghray
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Andrea Todisco
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Juanita L. Merchant
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
576
|
Nolan-Stevaux O, Lau J, Truitt ML, Chu GC, Hebrok M, Fernández-Zapico ME, Hanahan D. GLI1 is regulated through Smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. Genes Dev 2009; 23:24-36. [PMID: 19136624 DOI: 10.1101/gad.1753809] [Citation(s) in RCA: 321] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by the deregulation of the hedgehog signaling pathway. The Sonic Hedgehog ligand (Shh), absent in the normal pancreas, is highly expressed in pancreatic tumors and is sufficient to induce neoplastic precursor lesions in mouse models. We investigated the mechanism of Shh signaling in PDAC carcinogenesis by genetically ablating the canonical bottleneck of hedgehog signaling, the transmembrane protein Smoothened (Smo), in the pancreatic epithelium of PDAC-susceptible mice. We report that multistage development of PDAC tumors is not affected by the deletion of Smo in the pancreas, demonstrating that autocrine Shh-Ptch-Smo signaling is not required in pancreatic ductal cells for PDAC progression. However, the expression of Gli target genes is maintained in Smo-negative ducts, implicating alternative means of regulating Gli transcription in the neoplastic ductal epithelium. In PDAC tumor cells, we find that Gli transcription is decoupled from upstream Shh-Ptch-Smo signaling and is regulated by TGF-beta and KRAS, and we show that Gli1 is required both for survival and for the KRAS-mediated transformed phenotype of cultured PDAC cancer cells.
Collapse
Affiliation(s)
- Olivier Nolan-Stevaux
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94143, USA
| | | | | | | | | | | | | |
Collapse
|
577
|
Schrage YM, Hameetman L, Szuhai K, Cleton-Jansen AM, Taminiau AHM, Hogendoorn PCW, Bovée JVMG. Aberrant heparan sulfate proteoglycan localization, despite normal exostosin, in central chondrosarcoma. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:979-88. [PMID: 19179614 DOI: 10.2353/ajpath.2009.080623] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The tumor suppressor genes EXT1 and EXT2 are involved in the formation of multiple osteochondromas, which can progress to become secondary peripheral chondrosarcomas. The most common chondrosarcoma subtype is primary central chondrosarcoma, which occurs in the medullar cavity of bone. The EXT1/EXT2 protein complex is involved in heparan sulfate proteoglycan (HSPG) biosynthesis, which is important for signal transduction of Indian hedgehog (IHH), WNT, and transforming growth factor (TGF)-beta. The role of EXT and its downstream targets in central chondrosarcomas is currently unknown. EXT1 and EXT2 were therefore evaluated in central chondrosarcomas at both the DNA and mRNA levels. Immunohistochemistry was used to assess HSPG (CD44v3 and SDC2), WNT (beta-catenin), and TGF-beta (PAI-1 and phosphorylated Smad2) signaling, whereas IHH signaling was studied both by quantitative polymerase chain reaction and in vitro. mRNA levels of both EXT1 and EXT2 were normal in central chondrosarcomas; genomic alterations were absent in these regions and in 30 other HSPG-related genes. Although HSPGs were aberrantly located (CD44v3 in the Golgi and SDC2 in cytoplasm and nucleus), this was not caused by mutation. WNT signaling negatively correlated with increasing histological grade, whereas TGF-beta positively correlated with increasing histological grade. IHH signaling was active, and inhibition decreased cell viability in one of six cell lines. Our data suggest that, despite normal EXT in central chondrosarcomas, HSPGs and HSPG-dependent signaling are affected in both central and peripheral chondrosarcomas.
Collapse
Affiliation(s)
- Yvonne M Schrage
- Department of Pathology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
578
|
Cheng SY, Yue S. Role and regulation of human tumor suppressor SUFU in Hedgehog signaling. Adv Cancer Res 2009; 101:29-43. [PMID: 19055941 DOI: 10.1016/s0065-230x(08)00402-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Originally identified as factors affecting Drosophila embryogenesis, the Hedgehog (Hh) pathway is one of the primary signaling systems that specify patterns of cell growth and differentiation during vertebrate development. Mutations in various components of this pathway frequently occur in tumors originated from the skin, cerebellum, and skeletal muscle, and abnormal pathway activity is associated with a subset of lung, digestive tract, pancreatic, and prostate cancers. Because of these potent biological activities, this pathway is negatively regulated at multiple levels to ensure appropriate signaling responses. Suppressor of fused (Sufu) is one such negative regulator of Hh signaling. Although not essential in Drosophila, Sufu is absolutely required for mouse embryonic development. Mutations of Sufu are associated with a childhood brain tumor in human and an increased susceptibility to the same type of cancer in the TP53 null background in mice, and RNAi-mediated silencing of Sufu is sufficient to activate the Hh signaling in cultured fibroblasts. All these data point to a central role of Sufu in controlling the vertebrate Hh signaling pathway; however, for years what exactly Sufu does in the Hh pathway and what controls its activity remains a deep mystery. This chapter will go over all studies curated in the PubMed database with Sufu as a main subject during the past 17 years, and attempt to provide a balanced view on Sufu gene and protein structure, activities in Drosophila as well as mammalian development, and its involvement in cancer.
Collapse
Affiliation(s)
- Steven Y Cheng
- Center for Cancer Research, Nanjing Medical University , Nanjing, Jiangsu, PR China
| | | |
Collapse
|
579
|
Peukert S, Miller-Moslin K. Chapter 16 Hedgehog Signaling Pathway Inhibitors as Cancer Therapeutics. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2009. [DOI: 10.1016/s0065-7743(09)04416-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
580
|
Tsanev R, Tiigimägi P, Michelson P, Metsis M, Østerlund T, Kogerman P. Identification of the gene transcription repressor domain of Gli3. FEBS Lett 2008; 583:224-8. [PMID: 19084012 PMCID: PMC2697317 DOI: 10.1016/j.febslet.2008.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 11/27/2008] [Accepted: 12/04/2008] [Indexed: 01/20/2023]
Abstract
Gli transcription factors are downstream targets of the Hedgehog signaling pathway. Two of the three Gli proteins harbor gene transcription repressor function in the N-terminal half. We have analyzed the sequences and identified a potential repressor domain in Gli2 and Gli3 and have tested this experimentally. Overexpression studies confirm that the N-terminal parts harbor gene repression activity and we mapped the minimal repressor to residues 106 till 236 in Gli3. Unlike other mechanisms that inhibit Gli induced gene transcription, the repressor domain identified here does not utilize Histone deacetylases (HDACs) to achieve repression, as confirmed by HDAC inhibition studies and pull-down assays. This distinguishes the identified domain from other regulatory parts with negative influence on transcription.
Collapse
Affiliation(s)
- Robert Tsanev
- Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | | | | | | | | | | |
Collapse
|
581
|
Peukert S, Jain RK, Geisser A, Sun Y, Zhang R, Bourret A, Carlson A, Dasilva J, Ramamurthy A, Kelleher JF. Identification and structure-activity relationships of ortho-biphenyl carboxamides as potent Smoothened antagonists inhibiting the Hedgehog signaling pathway. Bioorg Med Chem Lett 2008; 19:328-31. [PMID: 19091559 DOI: 10.1016/j.bmcl.2008.11.096] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Revised: 11/24/2008] [Accepted: 11/24/2008] [Indexed: 11/20/2022]
Abstract
Ortho-biphenyl carboxamides, originally prepared as inhibitors of microsomal triglyceride transfer protein (MTP) have been identified as novel inhibitors of the Hedgehog signaling pathway. Structure-activity relationship studies for this class of compounds reduced MTP inhibitory activity and led to low nanomolar Hedgehog inhibitors. Binding assays revealed that the compounds act as antagonists of Smoothened and show cross-reactivity for both the human and mouse receptor.
Collapse
Affiliation(s)
- Stefan Peukert
- Novartis Institutes for BioMedical Research, Inc., 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
582
|
Ruat M, Angot E, Roudaut H, Traiffort E. Les antipsychotiques atypiques : revisiter les données pharmacologiques. Encephale 2008. [DOI: 10.1016/s0013-7006(08)75513-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
583
|
Abstract
The Hedgehog (Hh) pathway is one of the fundamental signal transduction pathways in animal development and is also involved in stem-cell maintenance and carcinogenesis. The hedgehog (hh) gene was first discovered in Drosophila, and members of the family have since been found in most metazoa. Hh proteins are composed of two domains, an amino-terminal domain HhN, which has the biological signal activity, and a carboxy-terminal autocatalytic domain HhC, which cleaves Hh into two parts in an intramolecular reaction and adds a cholesterol moiety to HhN. HhC has sequence similarity to the self-splicing inteins, and the shared region is termed Hint. New classes of proteins containing the Hint domain have been discovered recently in bacteria and eukaryotes, and the Hog class, of which Hh proteins comprise one family, is widespread throughout eukaryotes. The non-Hh Hog proteins have carboxy-terminal domains (the Hog domain) highly similar to HhC, although they lack the HhN domain, and instead have other amino-terminal domains. Hog proteins are found in many protists, but the Hh family emerged only in early metazoan evolution. HhN is modified by cholesterol at its carboxyl terminus and by palmitate at its amino terminus in both flies and mammals. The modified HhN is released from the cell and travels through the extracellular space. On binding its receptor Patched, it relieves the inhibition that Patched exerts on Smoothened, a G-protein-coupled receptor. The resulting signaling cascade converges on the transcription factor Cubitus interruptus (Ci), or its mammalian counterparts, the Gli proteins, which activate or repress target genes.
Collapse
Affiliation(s)
- Thomas R Bürglin
- Department of Biosciences and Nutrition, Karolinska Institutet, and School of Life Sciences, Södertörn University, Hälsovägen 7, SE-141 57 Huddinge, Sweden.
| |
Collapse
|
584
|
Arai MA, Tateno C, Hosoya T, Koyano T, Kowithayakorn T, Ishibashi M. Hedgehog/GLI-mediated transcriptional inhibitors from Zizyphus cambodiana. Bioorg Med Chem 2008; 16:9420-4. [DOI: 10.1016/j.bmc.2008.09.053] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 09/17/2008] [Accepted: 09/18/2008] [Indexed: 12/22/2022]
|
585
|
|
586
|
El-Sagheer AH, Brown T. Synthesis, Serum Stability and Cell Uptake of Cyclic and Hairpin Decoy Oligonucleotides for TCF/LEF and GLI Transcription Factors. Int J Pept Res Ther 2008. [DOI: 10.1007/s10989-008-9153-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
587
|
Chu D, Lu J. Novel therapies in breast cancer: what is new from ASCO 2008. J Hematol Oncol 2008; 1:16. [PMID: 18828924 PMCID: PMC2579406 DOI: 10.1186/1756-8722-1-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Accepted: 10/01/2008] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Breast cancer is the most common female cancer and the second most common cause of female cancer-related deaths in the United States. World-wide, more than one million women will be diagnosed with breast cancer annually. In 2007, more than 175,000 women were diagnosed with breast cancer in the United States. However, deaths due to breast cancer have decreased in the recent years in part because of improved screening techniques, surgical interventions, understanding of the pathogenesis of the disease, and utilization of traditional chemotherapies in a more efficacious manner. One of the more exciting areas of improvement in the treatment of breast cancer is the entrance of novel therapies now available to oncologists. In the field of cancer therapeutics, the area of targeted and biologic therapies has been progressing at a rapid rate, particularly in the treatment of breast cancer.Since the advent of imatinib for the successful treatment of chronic myelogenous leukemia in the 2001, clinicians have been searching for comparable therapies that could be as efficacious and as tolerable. In order for targeted therapies to be effective, the agent must be able to inhibit critical regulatory pathways which promote tumor cell growth and proliferation. The targets must be identifiable, quantifiable and capable of being interrupted.In the field of breast cancer, two advances in targeted therapy have led to great strides in the understanding and treatment of breast cancer, namely hormonal therapy for estrogen positive receptor breast cancer and antibodies directed towards the inhibition of human epidermal growth factor receptor (HER)2. These advances have revolutionized the understanding and the treatment strategies for breast cancer. Building upon these successes, a host of novel agents are currently being investigated and used in clinical trials that will hopefully prove to be as fruitful. This review will focus on novel therapies in the field of breast cancer with a focus on metastatic breast cancer (MBC) and updates from the recent annual ASCO meeting and contains a summary of the results.
Collapse
Affiliation(s)
- David Chu
- Division of Medical Oncology, Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Janice Lu
- Division of Medical Oncology, Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA
| |
Collapse
|
588
|
Wang Y, Price MA. A unique protection signal in Cubitus interruptus prevents its complete proteasomal degradation. Mol Cell Biol 2008; 28:5555-68. [PMID: 18625727 PMCID: PMC2546926 DOI: 10.1128/mcb.00524-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 04/30/2008] [Accepted: 07/02/2008] [Indexed: 11/20/2022] Open
Abstract
The limited proteolysis of Cubitus interruptus (Ci), the transcription factor for the developmentally and medically important Hedgehog (Hh) signaling pathway, triggers a critical switch between transcriptional repressor and activator forms. Ci repressor is formed when the C terminus of full-length Ci is degraded by the ubiquitin-proteasome pathway, an unusual reaction since the proteasome typically completely degrades its substrates. We show that several regions of Ci are required for generation of the repressor form: the zinc finger DNA binding domain, a single lysine residue (K750) near the degradation end point, and a 163-amino-acid region at the C terminus. Unlike other proteins that are partially degraded by the proteasome, dimerization is not a key feature of Ci processing. Using a pulse-chase assay in cultured Drosophila cells, we distinguish between regions required for initiation of degradation and those required for the protection of the Ci N terminus from degradation. We present a model whereby the zinc finger region and K750 together form a unique protection signal that prevents the complete degradation of Ci by the proteasome.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Biomedical Sciences, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | | |
Collapse
|
589
|
Yang SH, Andl T, Grachtchouk V, Wang A, Liu J, Syu LJ, Ferris J, Wang TS, Glick AB, Millar SE, Dlugosz AA. Pathological responses to oncogenic Hedgehog signaling in skin are dependent on canonical Wnt/beta3-catenin signaling. Nat Genet 2008; 40:1130-5. [PMID: 19165927 PMCID: PMC2688690 DOI: 10.1038/ng.192] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Constitutive Hedgehog (Hh) signaling underlies several human tumors, including basal cell carcinoma (BCC) and basaloid follicular hamartoma in skin. Intriguingly, superficial BCCs arise as de novo epithelial buds resembling embryonic hair germs, collections of epidermal cells whose development is regulated by canonical Wnt/beta-catenin signaling. Similar to embryonic hair germs, human BCC buds showed increased levels of cytoplasmic and nuclear beta-catenin and expressed early hair follicle lineage markers. We also detected canonical Wnt/ beta-catenin signaling in epithelial buds and hamartomas from mice expressing an oncogene, M2SMO, leading to constitutive Hh signaling in skin. Conditional overexpression of the Wnt pathway antagonist Dkk1 in M2SMO-expressing mice potently inhibited epithelial bud and hamartoma development without affecting Hh signaling. Our findings uncover a hitherto unknown requirement for ligand-driven, canonical Wnt/ beta-catenin signaling for Hh pathway-driven tumorigenesis, identify a new pharmacological target for these neoplasms and establish the molecular basis for the well-known similarity between early superficial BCCs and embryonic hair germs.
Collapse
Affiliation(s)
- Steven Hoseong Yang
- Department of Dermatology and Comprehensive Cancer Center, University of Michigan Medical School, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
590
|
Abstract
The hedgehog (Hh) pathway, initially discovered in Drosophila by two Nobel laureates, Dr. Eric Wieschaus and Dr. Christiane Nusslein-Volhard, is a major regulator for cell differentiation, tissue polarity and cell proliferation. Studies from many laboratories, including ours, reveal activation of this pathway in most basal cell carcinomas and in approximately 30% of extracutaneous human cancers, including medulloblastomas, gastrointestinal, lung, breast and prostate cancers. Thus, it is believed that targeted inhibition of Hh signaling may be effective in treating and preventing many types of human cancers. Even more exciting is the discovery and synthesis of specific signaling antagonists for the Hh pathway, which have significant clinical implications in novel cancer therapeutics. This review discusses the major advances in the current understanding of Hh signaling activation in different types of human cancers, the molecular basis of Hh signaling activation, the major antagonists for Hh signaling inhibition and their potential clinical application in human cancer therapy.
Collapse
Affiliation(s)
- Jingwu Xie
- Department of Pharmacology and Toxicology, Sealy Center for Cancer Cell Biology, University of Texas at Galveston, Galveston, Texas 77555-1048, USA.
| |
Collapse
|
591
|
Targeting of sonic hedgehog-GLI signaling: a potential strategy to improve therapy for mantle cell lymphoma. Mol Cancer Ther 2008; 7:1450-60. [DOI: 10.1158/1535-7163.mct-07-2118] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
592
|
Varjosalo M, Björklund M, Cheng F, Syvänen H, Kivioja T, Kilpinen S, Sun Z, Kallioniemi O, Stunnenberg HG, He WW, Ojala P, Taipale J. Application of active and kinase-deficient kinome collection for identification of kinases regulating hedgehog signaling. Cell 2008; 133:537-48. [PMID: 18455992 DOI: 10.1016/j.cell.2008.02.047] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 01/24/2008] [Accepted: 02/19/2008] [Indexed: 01/12/2023]
Abstract
To allow genome-scale identification of genes that regulate cellular signaling, we cloned >90% of all human full-length protein kinase cDNAs and constructed the corresponding kinase activity-deficient mutants. To establish the utility of this resource, we tested the effect of expression of the kinases on three different cellular signaling models. In all screens, many kinases had a modest but significant effect, apparently due to crosstalk between signaling pathways. However, the strongest effects were found with known regulators and novel components, such as MAP3K10 and DYRK2, which we identified in a mammalian Hedgehog (Hh) signaling screen. DYRK2 directly phosphorylated and induced the proteasome-dependent degradation of the key Hh pathway-regulated transcription factor, GLI2. MAP3K10, in turn, affected GLI2 indirectly by modulating the activity of DYRK2 and the known Hh pathway component, GSK3beta. Our results establish kinome expression screening as a highly effective way to identify physiological signaling pathway components and genes involved in pathological signaling crosstalk.
Collapse
Affiliation(s)
- Markku Varjosalo
- Department of Molecular Medicine, National Public Health Institute (KTL), FI00290 Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
593
|
Mimeault M, Hauke R, Batra SK. Recent advances on the molecular mechanisms involved in the drug resistance of cancer cells and novel targeting therapies. Clin Pharmacol Ther 2008; 83:673-91. [PMID: 17786164 PMCID: PMC2839198 DOI: 10.1038/sj.clpt.6100296] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review summarizes the recent knowledge obtained on the molecular mechanisms involved in the intrinsic and acquired resistance of cancer cells to current cancer therapies. We describe the cascades that are often altered in cancer cells during cancer progression that may contribute in a crucial manner to drug resistance and disease relapse. The emphasis is on the implication of ATP-binding cassette (ABC) multidrug efflux transporters in drug disposition and antiapoptotic factors, including epidermal growth factor receptor cascades and deregulated enzymes in ceramide metabolic pathways. The altered expression and activity of these signaling elements may have a critical role in the resistance of cancer cells to cytotoxic effects induced by diverse chemotherapeutic drugs and cancer recurrence. Of therapeutic interest, new strategies for reversing the multidrug resistance and developing more effective clinical treatments against the highly aggressive, metastatic, and recurrent cancers, based on the molecular targeting of the cancer progenitor cells and their further differentiated progeny, are also described.
Collapse
Affiliation(s)
- M Mimeault
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Eppley Institute of Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - R Hauke
- Eppley Institute of Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Division of Hematology and Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - SK Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Eppley Institute of Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
594
|
Mak KK, Bi Y, Wan C, Chuang PT, Clemens T, Young M, Yang Y. Hedgehog Signaling in Mature Osteoblasts Regulates Bone Formation and Resorption by Controlling PTHrP and RANKL Expression. Dev Cell 2008; 14:674-88. [DOI: 10.1016/j.devcel.2008.02.003] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 12/31/2007] [Accepted: 02/20/2008] [Indexed: 12/17/2022]
|
595
|
|
596
|
Bürglin TR. Evolution of hedgehog and hedgehog-related genes, their origin from Hog proteins in ancestral eukaryotes and discovery of a novel Hint motif. BMC Genomics 2008; 9:127. [PMID: 18334026 PMCID: PMC2362128 DOI: 10.1186/1471-2164-9-127] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Accepted: 03/11/2008] [Indexed: 11/18/2022] Open
Abstract
Background The Hedgehog (Hh) signaling pathway plays important roles in human and animal development as well as in carcinogenesis. Hh molecules have been found in both protostomes and deuterostomes, but curiously the nematode Caenorhabditis elegans lacks a bona-fide Hh. Instead a series of Hh-related proteins are found, which share the Hint/Hog domain with Hh, but have distinct N-termini. Results We performed extensive genome searches such as the cnidarian Nematostella vectensis and several nematodes to gain further insights into Hh evolution. We found six genes in N. vectensis with a relationship to Hh: two Hh genes, one gene with a Hh N-terminal domain fused to a Willebrand factor type A domain (VWA), and three genes containing Hint/Hog domains with distinct novel N-termini. In the nematode Brugia malayi we find the same types of hh-related genes as in C. elegans. In the more distantly related Enoplea nematodes Xiphinema and Trichinella spiralis we find a bona-fide Hh. In addition, T. spiralis also has a quahog gene like C. elegans, and there are several additional hh-related genes, some of which have secreted N-terminal domains of only 15 to 25 residues. Examination of other Hh pathway components revealed that T. spiralis - like C. elegans - lacks some of these components. Extending our search to all eukaryotes, we recovered genes containing a Hog domain similar to Hh from many different groups of protists. In addition, we identified a novel Hint gene family present in many eukaryote groups that encodes a VWA domain fused to a distinct Hint domain we call Vint. Further members of a poorly characterized Hint family were also retrieved from bacteria. Conclusion In Cnidaria and nematodes the evolution of hh genes occurred in parallel to the evolution of other genes that contain a Hog domain but have different N-termini. The fact that Hog genes comprising a secreted N-terminus and a Hog domain are found in many protists indicates that this gene family must have arisen in very early eukaryotic evolution, and gave rise eventually to hh and hh-related genes in animals. The results indicate a hitherto unsuspected ability of Hog domain encoding genes to evolve new N-termini. In one instance in Cnidaria, the Hh N-terminal signaling domain is associated with a VWA domain and lacks a Hog domain, suggesting a modular mode of evolution also for the N-terminal domain. The Hog domain proteins, the inteins and VWA-Vint proteins are three families of Hint domain proteins that evolved in parallel in eukaryotes.
Collapse
Affiliation(s)
- Thomas R Bürglin
- Dept. of Biosciences and Nutrition, Karolinska Institutet & School of Life Sciences, Södertörns Högskola, Alfred Nobels Allé 7, SE-141 89 Huddinge, Sweden.
| |
Collapse
|
597
|
Abstract
Hedgehog-mediated signaling has been shown to promote growth and dissemination of solid cancers, most prominently basal cell carcinomas and medulloblastoma. Recent findings indicate that hedgehog signals are also important for tumor growth in hematologic malignancies. Hedgehog ligands secreted by stromal cells could elicit Patched/Smoothened-mediated antiapoptotic signaling in mouse B-cell lymphomas. Inhibition of hedgehog signaling induced apoptosis in lymphoma cells and prolonged survival of lymphoma-bearing mice. Depletion of tumor cells proceeded in the absence of p53 via the mitochondrial apoptotic pathway. These and other recently published data on hedgehog inhibition in cancer cells and their implications will be discussed.
Collapse
Affiliation(s)
- Ralph K Lindemann
- National Center for Tumor Diseases, Department of Translational Oncology G100, Heidelberg, Germany.
| |
Collapse
|
598
|
Fan Y, Bergmann A. Distinct mechanisms of apoptosis-induced compensatory proliferation in proliferating and differentiating tissues in the Drosophila eye. Dev Cell 2008; 14:399-410. [PMID: 18331718 PMCID: PMC2277325 DOI: 10.1016/j.devcel.2008.01.003] [Citation(s) in RCA: 193] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Revised: 10/15/2007] [Accepted: 01/07/2008] [Indexed: 01/08/2023]
Abstract
In multicellular organisms, apoptotic cells induce compensatory proliferation of neighboring cells to maintain tissue homeostasis. In the Drosophila wing imaginal disc, dying cells trigger compensatory proliferation through secretion of the mitogens Decapentaplegic (Dpp) and Wingless (Wg). This process is under control of the initiator caspase Dronc, but not effector caspases. Here we show that a second mechanism of apoptosis-induced compensatory proliferation exists. This mechanism is dependent on effector caspases which trigger the activation of Hedgehog (Hh) signaling for compensatory proliferation. Furthermore, whereas Dpp and Wg signaling is preferentially employed in apoptotic proliferating tissues, Hh signaling is activated in differentiating eye tissues. Interestingly, effector caspases in photoreceptor neurons stimulate Hh signaling which triggers cell-cycle reentry of cells that had previously exited the cell cycle. In summary, dependent on the developmental potential of the affected tissue, different caspases trigger distinct forms of compensatory proliferation in an apparent nonapoptotic function.
Collapse
Affiliation(s)
- Yun Fan
- Department of Biochemistry & Molecular Biology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard – Unit 1000, Houston, TX 77030
| | - Andreas Bergmann
- Department of Biochemistry & Molecular Biology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard – Unit 1000, Houston, TX 77030
| |
Collapse
|
599
|
Rudin CM, Hann CL, Peacock CD, Watkins DN. Novel systemic therapies for small cell lung cancer. J Natl Compr Canc Netw 2008; 6:315-22. [PMID: 18377849 PMCID: PMC4086469 DOI: 10.6004/jnccn.2008.0026] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Accepted: 11/16/2007] [Indexed: 02/06/2023]
Abstract
A diagnosis of small cell lung cancer (SCLC) today confers essentially the same terrible prognosis that it did 25 years ago, when common use of cisplatin-based chemotherapy began for this disease. In contrast to past decades of research on many other solid tumors, studies of combination chemotherapy using later generation cytotoxics and targeted kinase inhibitors have not had a significant impact on standard care for SCLC. The past few years have seen suggestions of incrementally improved outcomes using standard cytotoxics, including cisplatin-based combination studies of irinotecan and amrubicin by Japanese research consortia. Confirmatory phase III studies of these agents are ongoing in the United States. Antiangiogenic strategies are also of primary interest and are in late-phase testing. Several novel therapeutics, including high-potency small molecule inhibitors of Bcl-2 and the Hedgehog signaling pathway, and a recently discovered replication-competent picornavirus, have shown remarkable activity against SCLC in preclinical models and are currently in simultaneous phase I clinical development. Novel therapeutic approaches based on advances in understanding of the biology of SCLC have the potential to radically change the outlook for patients with this disease.
Collapse
Affiliation(s)
- Charles M Rudin
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231, USA.
| | | | | | | |
Collapse
|
600
|
Kimura H, Ng JMY, Curran T. Transient inhibition of the Hedgehog pathway in young mice causes permanent defects in bone structure. Cancer Cell 2008; 13:249-60. [PMID: 18328428 DOI: 10.1016/j.ccr.2008.01.027] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 11/20/2007] [Accepted: 01/14/2008] [Indexed: 10/22/2022]
Abstract
The Hedgehog (Hh) pathway plays critical roles in normal development and in tumorigenesis. We generated Gli-luciferase transgenic mice to evaluate the Smo inhibitor, HhAntag, by whole animal functional imaging. HhAntag rapidly reduced systemic luciferase activity in 10- to 14-day-old mice following oral dosing. Although pathway activity was restored 2 days after drug removal, brief inhibition caused permanent defects in bone growth. HhAntag inhibited proliferation and promoted differentiation of chondrocytes, leading to dramatic expansion of the hypertrophic zone. After drug removal, osteoblasts invaded the cartilage plate, mineralization occurred, and there was premature fusion of the growth plate resulting in permanent disruption of bone epiphyses.
Collapse
MESH Headings
- Administration, Oral
- Aging/metabolism
- Animals
- Animals, Newborn
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/toxicity
- Bone Remodeling/drug effects
- Bone and Bones/drug effects
- Bone and Bones/embryology
- Bone and Bones/metabolism
- Bone and Bones/pathology
- Calcification, Physiologic/drug effects
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cerebellar Neoplasms/drug therapy
- Chondrocytes/drug effects
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Dose-Response Relationship, Drug
- Growth Plate/drug effects
- Growth Plate/pathology
- Hedgehog Proteins/metabolism
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Luciferases/genetics
- Luciferases/metabolism
- Medulloblastoma/drug therapy
- Mice
- Mice, Transgenic
- Microscopy, Fluorescence
- Microscopy, Video
- Osteogenesis/drug effects
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/metabolism
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/drug effects
- Smoothened Receptor
- Time Factors
- Zinc Finger Protein GLI1
Collapse
Affiliation(s)
- Hiromichi Kimura
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 332 North Lauderdale Street, Memphis, TN 38105, USA
| | | | | |
Collapse
|