1
|
Müller GA, Müller TD. (Patho)Physiology of Glycosylphosphatidylinositol-Anchored Proteins I: Localization at Plasma Membranes and Extracellular Compartments. Biomolecules 2023; 13:biom13050855. [PMID: 37238725 DOI: 10.3390/biom13050855] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (APs) are anchored at the outer leaflet of plasma membranes (PMs) of all eukaryotic organisms studied so far by covalent linkage to a highly conserved glycolipid rather than a transmembrane domain. Since their first description, experimental data have been accumulating for the capability of GPI-APs to be released from PMs into the surrounding milieu. It became evident that this release results in distinct arrangements of GPI-APs which are compatible with the aqueous milieu upon loss of their GPI anchor by (proteolytic or lipolytic) cleavage or in the course of shielding of the full-length GPI anchor by incorporation into extracellular vesicles, lipoprotein-like particles and (lyso)phospholipid- and cholesterol-harboring micelle-like complexes or by association with GPI-binding proteins or/and other full-length GPI-APs. In mammalian organisms, the (patho)physiological roles of the released GPI-APs in the extracellular environment, such as blood and tissue cells, depend on the molecular mechanisms of their release as well as the cell types and tissues involved, and are controlled by their removal from circulation. This is accomplished by endocytic uptake by liver cells and/or degradation by GPI-specific phospholipase D in order to bypass potential unwanted effects of the released GPI-APs or their transfer from the releasing donor to acceptor cells (which will be reviewed in a forthcoming manuscript).
Collapse
Affiliation(s)
- Günter A Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), 85764 Oberschleissheim, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764 Oberschleissheim, Germany
- German Center for Diabetes Research (DZD), 85764 Oberschleissheim, Germany
| |
Collapse
|
2
|
Dai OL, Lei ZY, Peng YD, Wang Z. Integrative analysis uncovers response mechanism of Pirata subpiraticus to chronic cadmium stress. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:90070-90080. [PMID: 35864398 DOI: 10.1007/s11356-022-22043-7] [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: 04/05/2022] [Accepted: 07/12/2022] [Indexed: 06/15/2023]
Abstract
Soil cadmium (Cd) pollution is global environmental pollution and adversely affects paddy field organisms. Wolf spider grants a new insight to evaluate the toxicity triggered by Cd, yet the impact of chronic Cd exposure on the spider and its molecular mechanism remains unclear. The present study found that the wolf spider Pirata subpiraticus fed with Cd-accumulated flies for 5 weeks presented lower catalase, peroxidase, and acetylcholinesterase activities and higher malonaldehyde content than the control spiders (p < 0.05). An in-depth transcriptomic analysis yielded a total of 5995 differentially expressed genes (DEGs, with 3857 up-regulated and 2138 down-regulated genes) from the comparison, and 19 DEGs encoding three enzymatic indicators were down-regulated. Further enrichment analysis indicated that Cd stress could inhibit the expression of cuticle and chitin-encoding genes via the down-regulation of several key enzymes, such as chitin synthase, glutamine-fructose-6-phosphate transaminase, and chitinase. In addition, our findings suggested that hedgehog and FoxO signaling pathways might play an essential role in regulating survival, cell cycle, and autophagy process in spiders, which were primarily down-regulated under Cd stress. An intensely interactive network displayed that Cd exposure could repress key biological processes in P. subpiraticus, particularly peptide metabolic process and peptide biosynthetic process. To sum up, this integrative investigation confirmed an effective bioindicator for assessing Cd-induced toxicity; provided a mass of genes, proteins, and enzymes for further validation; and granted novel perspectives to uncover the molecular responses of spiders to Cd pollution.
Collapse
Affiliation(s)
- Ou-Lin Dai
- College of Resources and Environment, Hunan Agricultural University, Changsha, 410128, China
| | - Zi-Yan Lei
- College of Resources and Environment, Hunan Agricultural University, Changsha, 410128, China
| | - Yuan-de Peng
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, 410205, China
| | - Zhi Wang
- College of Resources and Environment, Hunan Agricultural University, Changsha, 410128, China.
- College of Life Science, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
3
|
Strilbytska OM, Semaniuk UV, Strutynska TR, Burdyliuk NI, Tsiumpala S, Bubalo V, Lushchak O. Herbicide Roundup shows toxic effects in nontarget organism Drosophila. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 110:e21893. [PMID: 35388481 DOI: 10.1002/arch.21893] [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: 02/04/2022] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 06/14/2023]
Abstract
Glyphosate-based herbicide Roundup, as the most employed herbicide used for multiple purposes in agriculture, adversely affects nontarget organisms. We tested the effects of Roundup applied at larval and adult stages. Roundup caused developmental delay and increased larvae mortality. Roundup treatment reduced hemolymph glucose and glycogen levels in adult flies of both sexes at the highest concentration tested. Sex-dependent diverse effects were found in catalase and Cu,Zn superoxide dismutase (Cu,Zn-SOD) activities. Decreased aconitase activity, contents of thiols, and lipid peroxides were found after larval Roundup exposure. Furthermore, chronic exposure to adult flies decreased appetite, body weight, and shortened lifespan. Thus, our results suggest that high concentrations of Roundup are deleterious to both larvae and adults, resulting in a shift of the metabolism and antioxidant defense system in Drosophila melanogaster.
Collapse
Affiliation(s)
- Olha M Strilbytska
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Uliana V Semaniuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Tetiana R Strutynska
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Nadia I Burdyliuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Sviatoslav Tsiumpala
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Volodymyr Bubalo
- Laboratory of Experimental Toxicology and Mutagenesis, L.I. Medved's Research Center of Preventive Toxicology, Food and Chemical Safety, MHU, Kyiv, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
4
|
Garg C, khan H, Kaur A, Singh TG, Sharma VK, Singh SK. Therapeutic Implications of Sonic Hedgehog Pathway in Metabolic Disorders: Novel Target for Effective Treatment. Pharmacol Res 2022; 179:106194. [DOI: 10.1016/j.phrs.2022.106194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 12/13/2022]
|
5
|
Peer E, Aichberger SK, Vilotic F, Gruber W, Parigger T, Grund-Gröschke S, Elmer DP, Rathje F, Ramspacher A, Zaja M, Michel S, Hamm S, Aberger F. Casein Kinase 1D Encodes a Novel Drug Target in Hedgehog-GLI-Driven Cancers and Tumor-Initiating Cells Resistant to SMO Inhibition. Cancers (Basel) 2021; 13:cancers13164227. [PMID: 34439381 PMCID: PMC8394935 DOI: 10.3390/cancers13164227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Uncontrolled activation of hedgehog (HH)—GLI signaling contributes to the development of several human malignancies. Targeted inhibition of the HH—GLI signaling cascade with small-molecule inhibitors can reduce cancer growth, but patient relapse is very common due to the development of drug resistance. Therefore, a high unmet medical need exists for new drug targets and inhibitors to achieve efficient and durable responses. In the current study, we identified CSNK1D as a novel drug target in the HH—GLI signaling pathway. Genetic and pharmacological inhibition of CSNK1D activity leads to suppression of oncogenic HH—GLI signaling, even in cancer cells in which already approved HH inhibitors are no longer effective due to resistance mechanisms. Inhibition of CSNK1D function reduces the malignant properties of so-called tumor-initiating cells, thereby limiting cancer growth and presumably metastasis. The results of this study form the basis for the development of efficient CSNK1D inhibitors for the therapy of HH—GLI-associated cancers. Abstract (1) Background: Aberrant activation of the hedgehog (HH)—GLI pathway in stem-like tumor-initiating cells (TIC) is a frequent oncogenic driver signal in various human malignancies. Remarkable efficacy of anti-HH therapeutics led to the approval of HH inhibitors targeting the key pathway effector smoothened (SMO) in basal cell carcinoma and acute myeloid leukemia. However, frequent development of drug resistance and severe adverse effects of SMO inhibitors pose major challenges that require alternative treatment strategies targeting HH—GLI in TIC downstream of SMO. We therefore investigated members of the casein kinase 1 (CSNK1) family as novel drug targets in HH—GLI-driven malignancies. (2) Methods: We genetically and pharmacologically inhibited CSNK1D in HH-dependent cancer cells displaying either sensitivity or resistance to SMO inhibitors. To address the role of CSNK1D in oncogenic HH signaling and tumor growth and initiation, we quantitatively analyzed HH target gene expression, performed genetic and chemical perturbations of CSNK1D activity, and monitored the oncogenic transformation of TIC in vitro and in vivo using 3D clonogenic tumor spheroid assays and xenograft models. (3) Results: We show that CSNK1D plays a critical role in controlling oncogenic GLI activity downstream of SMO. We provide evidence that inhibition of CSNK1D interferes with oncogenic HH signaling in both SMO inhibitor-sensitive and -resistant tumor settings. Furthermore, genetic and pharmacologic perturbation of CSNK1D decreases the clonogenic growth of GLI-dependent TIC in vitro and in vivo. (4) Conclusions: Pharmacologic targeting of CSNK1D represents a novel therapeutic approach for the treatment of both SMO inhibitor-sensitive and -resistant tumors.
Collapse
Affiliation(s)
- Elisabeth Peer
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
| | - Sophie Karoline Aichberger
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
| | - Filip Vilotic
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
| | - Wolfgang Gruber
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
| | - Thomas Parigger
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
- Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Salzburg Cancer Research Institute, Cancer Cluster Salzburg, IIIrd Medical Department, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Sandra Grund-Gröschke
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
| | - Dominik Patrick Elmer
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
| | - Florian Rathje
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
| | - Andrea Ramspacher
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mirko Zaja
- 4SC AG, Planegg-Martinsried, 82152 Planegg, Germany; (M.Z.); (S.M.); (S.H.)
| | - Susanne Michel
- 4SC AG, Planegg-Martinsried, 82152 Planegg, Germany; (M.Z.); (S.M.); (S.H.)
| | - Svetlana Hamm
- 4SC AG, Planegg-Martinsried, 82152 Planegg, Germany; (M.Z.); (S.M.); (S.H.)
| | - Fritz Aberger
- Department of Bioscience, Cancer Cluster Salzburg, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (E.P.); (S.K.A.); (F.V.); (W.G.); (T.P.); (S.G.-G.); (D.P.E.); (F.R.); (A.R.)
- Correspondence: ; Tel.: +43-662-8044-5792
| |
Collapse
|
6
|
Morival JLP, Widyastuti HP, Nguyen CHH, Zaragoza MV, Downing TL. DNA methylation analysis reveals epimutation hotspots in patients with dilated cardiomyopathy-associated laminopathies. Clin Epigenetics 2021; 13:139. [PMID: 34246298 PMCID: PMC8272901 DOI: 10.1186/s13148-021-01127-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/03/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Mutations in LMNA, encoding lamin A/C, lead to a variety of diseases known as laminopathies including dilated cardiomyopathy (DCM) and skeletal abnormalities. Though previous studies have investigated the dysregulation of gene expression in cells from patients with DCM, the role of epigenetic (gene regulatory) mechanisms, such as DNA methylation, has not been thoroughly investigated. Furthermore, the impact of family-specific LMNA mutations on DNA methylation is unknown. Here, we performed reduced representation bisulfite sequencing on ten pairs of fibroblasts and their induced pluripotent stem cell (iPSC) derivatives from two families with DCM due to distinct LMNA mutations, one of which also induces brachydactyly. RESULTS Family-specific differentially methylated regions (DMRs) were identified by comparing the DNA methylation landscape of patient and control samples. Fibroblast DMRs were found to enrich for distal regulatory features and transcriptionally repressed chromatin and to associate with genes related to phenotypes found in tissues affected by laminopathies. These DMRs, in combination with transcriptome-wide expression data and lamina-associated domain (LAD) organization, revealed the presence of inter-family epimutation hotspots near differentially expressed genes, most of which were located outside LADs redistributed in LMNA-related DCM. Comparison of DMRs found in fibroblasts and iPSCs identified regions where epimutations were persistent across both cell types. Finally, a network of aberrantly methylated disease-associated genes revealed a potential molecular link between pathways involved in bone and heart development. CONCLUSIONS Our results identified both shared and mutation-specific laminopathy epimutation landscapes that were consistent with lamin A/C mutation-mediated epigenetic aberrancies that arose in somatic and early developmental cell stages.
Collapse
Affiliation(s)
- Julien L. P. Morival
- Department of Biomedical Engineering and The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, 2408 Engineering III, Irvine, CA 92697 USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California Irvine, Irvine, CA USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA USA
| | - Halida P. Widyastuti
- UCI Cardiogenomics Program, Department of Pediatrics, Division of Genetics and Genomics and Department of Biological Chemistry, University of California Irvine, 2042 Hewitt Hall, Irvine, CA 92697 USA
| | - Cecilia H. H. Nguyen
- UCI Cardiogenomics Program, Department of Pediatrics, Division of Genetics and Genomics and Department of Biological Chemistry, University of California Irvine, 2042 Hewitt Hall, Irvine, CA 92697 USA
| | - Michael V. Zaragoza
- UCI Cardiogenomics Program, Department of Pediatrics, Division of Genetics and Genomics and Department of Biological Chemistry, University of California Irvine, 2042 Hewitt Hall, Irvine, CA 92697 USA
| | - Timothy L. Downing
- Department of Biomedical Engineering and The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, 2408 Engineering III, Irvine, CA 92697 USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California Irvine, Irvine, CA USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA USA
- Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, CA USA
| |
Collapse
|
7
|
Cui G, Yuan H, Jiang Z, Zhang J, Sun Z, Zhong G. Natural harmine negatively regulates the developmental signaling network of Drosophila melanogaster (Drosophilidae: Diptera) in vivo. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 190:110134. [PMID: 31901541 DOI: 10.1016/j.ecoenv.2019.110134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 06/10/2023]
Abstract
The widely distributed β-carboline alkaloids exhibit promising psychopharmacological and biochemical effects. Harmine, a natural β-carboline, can inhibit insect growth and development with unclear mechanisms. In this study, harmine (at 0-200 mg/L) showed a dose-dependent inhibitory effect on the pupal weight, length, height, pupation rate and eclosion rate of fruit flies Drosophila melanogaster, which was similar to the inhibition induced by the well-known botanical insect growth regulator azadirachtin. Moreover, the expression levels of major regulators from the developmental signaling network were down-regulated during the pupal stage except Numb, Fringe, Yorkie and Pten. The Notch, Wnt, Hedgehog and TGF-β pathways mainly played vital roles in coping with harmine exposure in pupae stage, while the Hippo, Hedgehog and TGF-β elements were involved in the sex differences. Notch, Hippo, Hedgehog, Dpp and Armadillo were proved to be suppressed in the developmental inhibition with fly mutants, while Numb and Punt were increased by harmine. In conclusion, harmine significantly inhibited the development of Drosophila by negatively affecting their developmental signaling network during different stages. Our results establish a preliminary understanding of the developmental signaling network subjected to botanical component-induced growth inhibition and lay the groundwork for further application.
Collapse
Affiliation(s)
- Gaofeng Cui
- Key Laboratory of Integrated Pest Management on Crops in South China, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Natural Pesticide & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, 510642, China.
| | - Haiqi Yuan
- Key Laboratory of Integrated Pest Management on Crops in South China, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Natural Pesticide & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, 510642, China.
| | - Zhiyan Jiang
- Key Laboratory of Integrated Pest Management on Crops in South China, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Natural Pesticide & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, 510642, China.
| | - Jing Zhang
- Key Laboratory of Integrated Pest Management on Crops in South China, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Natural Pesticide & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, 510642, China.
| | - Zhipeng Sun
- Key Laboratory of Integrated Pest Management on Crops in South China, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Natural Pesticide & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, 510642, China.
| | - Guohua Zhong
- Key Laboratory of Integrated Pest Management on Crops in South China, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China; Key Laboratory of Natural Pesticide & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
8
|
Lentivirus-mediated silencing of the PTC1 and PTC2 genes promotes recovery from spinal cord injury by activating the Hedgehog signaling pathway in a rat model. Exp Mol Med 2017; 49:e412. [PMID: 29244790 PMCID: PMC5750477 DOI: 10.1038/emm.2017.220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 06/02/2017] [Accepted: 06/25/2017] [Indexed: 12/29/2022] Open
Abstract
This study aimed to investigate the effect of Patched-1 (PTC1) and PTC2 silencing in a rat model, on Hedgehog (Hh) pathway-mediated recovery from spinal cord injury (SCI). An analytical emphasis on the relationship between the sonic hedgehog (Shh) pathway and nerve regeneration was explored. A total of 126 rats were divided into normal, sham, SCI, negative control (NC), PTC1-RNAi, PTC2-RNAi and PTC1/PTC2-RNAi groups. The Basso, Beattie and Bresnahan (BBB) scale was employed to assess hind limb motor function. Quantitative real-time polymerase chain reaction and western blotting were performed to examine the mRNA and protein levels of PTC1, PTC2, Shh, glioma-associated oncogene homolog 1 (Gli-1), Smo and Nestin. Tissue morphology was analyzed using immunohistochemistry, and immunofluorescent staining was conducted to detect neurofilament protein 200 (NF-200) and glial fibrillary acidic protein (GFAP). The PTC1/PTC2-RNAi group displayed higher BBB scores than the SCI and NC groups. Shh, Gli-1, Smo and Nestin expression levels were elevated in the PTC1/PTC2-RNAi group. PTC1 and PTC2 mRNA and protein expression was lower in the PTC1/PTC2-RNAi group than in the normal, sham and SCI groups. Among the seven groups, the PTC1/PTC2-RNAi group had the largest positive area of NF-200 staining, whereas the SCI group exhibited a larger GFAP-positive area than both the normal and the sham groups. The Shh pathway may provide new insights into therapeutic indications and regenerative recovery tools for the treatment of SCI. Activation of the Hh signaling pathway by silencing PTC1 and PTC2 may reduce inflammation and may ultimately promote SCI recovery.
Collapse
|
9
|
Im SH, Takle K, Jo J, Babcock DT, Ma Z, Xiang Y, Galko MJ. Tachykinin acts upstream of autocrine Hedgehog signaling during nociceptive sensitization in Drosophila. eLife 2015; 4:e10735. [PMID: 26575288 PMCID: PMC4739760 DOI: 10.7554/elife.10735] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022] Open
Abstract
Pain signaling in vertebrates is modulated by neuropeptides like Substance P (SP). To determine whether such modulation is conserved and potentially uncover novel interactions between nociceptive signaling pathways we examined SP/Tachykinin signaling in a Drosophila model of tissue damage-induced nociceptive hypersensitivity. Tissue-specific knockdowns and genetic mutant analyses revealed that both Tachykinin and Tachykinin-like receptor (DTKR99D) are required for damage-induced thermal nociceptive sensitization. Electrophysiological recording showed that DTKR99D is required in nociceptive sensory neurons for temperature-dependent increases in firing frequency upon tissue damage. DTKR overexpression caused both behavioral and electrophysiological thermal nociceptive hypersensitivity. Hedgehog, another key regulator of nociceptive sensitization, was produced by nociceptive sensory neurons following tissue damage. Surprisingly, genetic epistasis analysis revealed that DTKR function was upstream of Hedgehog-dependent sensitization in nociceptive sensory neurons. Our results highlight a conserved role for Tachykinin signaling in regulating nociception and the power of Drosophila for genetic dissection of nociception. DOI:http://dx.doi.org/10.7554/eLife.10735.001 Injured animals from humans to insects become extra sensitive to sensations such as touch and heat. This hypersensitivity is thought to protect areas of injury or inflammation while they heal, but it is not clear how it comes about. Now, Im et al. have addressed this question by assessing pain in fruit flies after tissue damage. The experiments used ultraviolet radiation to essentially cause ‘localized sunburn’ to fruit fly larvae. Electrical impulses were then recorded from the larvae’s pain-detecting neurons and the larvae were analyzed for behaviors that indicate pain responses (for example, rolling). Im et al. found that tissue injury lowers the threshold at which temperature causes pain in fruit fly larvae. Further experiments using mutant flies that lacked genes involved in two signaling pathways showed that a signaling molecule called Tachykinin and its receptor (called DTKR) are needed to regulate the observed threshold lowering. When the genes for either of these proteins were deleted, the larvae no longer showed the pain hypersensitivity following an injury. Further experiments then uncovered a genetic interaction between Tachykinin signaling and a second signaling pathway that also regulates pain sensitization (called Hedgehog signaling). Im et al. found that Tachykinin acts upstream of Hedgehog in the pain-detecting neurons. Following on from these findings, the biggest outstanding questions are: how, when and where does tissue damage lead to the release of Tachykinin to sensitize neurons? Future studies could also ask whether the genetic interactions between Hedgehog and Tachykinin (or related proteins) are conserved in other animals such as humans and mice. DOI:http://dx.doi.org/10.7554/eLife.10735.002
Collapse
Affiliation(s)
- Seol Hee Im
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, United States
| | - Kendra Takle
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Juyeon Jo
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, United States.,Genes and Development Graduate Program, University of Texas Graduate School of Biomedical Sciences, Houston, United States
| | - Daniel T Babcock
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, United States.,Neuroscience Graduate Program, University of Texas Graduate School of Biomedical Sciences, Houston, United States
| | - Zhiguo Ma
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Yang Xiang
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, United States
| | - Michael J Galko
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, United States.,Genes and Development Graduate Program, University of Texas Graduate School of Biomedical Sciences, Houston, United States.,Neuroscience Graduate Program, University of Texas Graduate School of Biomedical Sciences, Houston, United States
| |
Collapse
|
10
|
Cheseto X, Kuate SP, Tchouassi DP, Ndung’u M, Teal PEA, Torto B. Potential of the Desert Locust Schistocerca gregaria (Orthoptera: Acrididae) as an Unconventional Source of Dietary and Therapeutic Sterols. PLoS One 2015; 10:e0127171. [PMID: 25970517 PMCID: PMC4429980 DOI: 10.1371/journal.pone.0127171] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/12/2015] [Indexed: 11/19/2022] Open
Abstract
Insects are increasingly being recognized not only as a source of food to feed the ever growing world population but also as potential sources of new products and therapeutic agents, among which are sterols. In this study, we sought to profile sterols and their derivatives present in the desert locust, Schistocerca gregaria, focusing on those with potential importance as dietary and therapeutic components for humans. Using coupled gas chromatography-mass spectrometry (GC-MS), we analyzed and compared the quantities of sterols in the different sections of the gut and tissues of the locust. In the gut, we identified 34 sterols which showed a patchy distribution, but with the highest composition in the foregut (55%) followed by midgut (31%) and hindgut (14%). Fed ad libitum on wheat seedlings, five sterols unique to the insect were detected. These sterols were identified as 7-dehydrocholesterol, desmosterol, fucosterol, (3β, 5α) cholesta-8, 14, 24-trien-3-ol, 4, 4-dimethyl, and (3β, 20R) cholesta-5, 24-dien-3, 20-diol with the first three having known health benefits in humans. Incubation of the fore-, mid- and hindgut with cholesterol-[4-13C] yielded eight derivatives, three of these were detected in the gut of the desert locust after it had consumed the vegetative diet but were not detected in the diet. Our study shows that the desert locust ingests phytosterols from a vegetative diet and, amplifies and metabolizes them into derivatives with potential salutary benefits and we discuss our findings in this context.
Collapse
Affiliation(s)
- Xavier Cheseto
- Behavioral and Chemical Ecology Department, International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Chemistry Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Serge Philibert Kuate
- Behavioral and Chemical Ecology Department, International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - David P. Tchouassi
- Behavioral and Chemical Ecology Department, International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Mary Ndung’u
- Chemistry Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Peter E. A. Teal
- Chemistry Research Unit, Center for Medical, Agricultural, and Veterinary Entomology, United States Department of Agriculture/Agricultural Research Service, Gainesville, Florida, United States of America
| | - Baldwyn Torto
- Behavioral and Chemical Ecology Department, International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- * E-mail:
| |
Collapse
|
11
|
Grubbs N, Leach M, Su X, Petrisko T, Rosario JB, Mahaffey JW. New components of Drosophila leg development identified through genome wide association studies. PLoS One 2013; 8:e60261. [PMID: 23560084 PMCID: PMC3613359 DOI: 10.1371/journal.pone.0060261] [Citation(s) in RCA: 12] [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: 12/04/2012] [Accepted: 02/24/2013] [Indexed: 11/29/2022] Open
Abstract
The adult Drosophila melanogaster body develops from imaginal discs, groups of cells set-aside during embryogenesis and expanded in number during larval stages. Specification and development of Drosophila imaginal discs have been studied for many years as models of morphogenesis. These studies are often based on mutations with large developmental effects, mutations that are often lethal in embryos when homozygous. Such forward genetic screens can be limited by factors such as early lethality and genetic redundancy. To identify additional genes and genetic pathways involved in leg imaginal disc development, we employed a Genome Wide Association Study utilizing the natural genetic variation in leg proportionality found in the Drosophila Genetic Reference Panel fly lines. In addition to identifying genes already known to be involved in leg development, we identified several genes involved in pathways that had not previously been linked with leg development. Several of the genes appear to be involved in signaling activities, while others have no known roles at this time. Many of these uncharacterized genes are conserved in mammals, so we can now begin to place these genes into developmental contexts. Interestingly, we identified five genes which, when their function is reduced by RNAi, cause an antenna-to-leg transformation. Our results demonstrate the utility of this approach, integrating the tools of quantitative and molecular genetics to study developmental processes, and provide new insights into the pathways and networks involved in Drosophila leg development.
Collapse
Affiliation(s)
- Nathaniel Grubbs
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Megan Leach
- Department of Plant Pathology, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Xin Su
- Transgenics Department, Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
| | | | - Juan B. Rosario
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - James W. Mahaffey
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
12
|
Cho ES, Lim SS, Hwang JW, Lee JC. Constitutive activation of smoothened leads to impaired developments of postnatal bone in mice. Mol Cells 2012; 34:399-405. [PMID: 22983747 PMCID: PMC3887766 DOI: 10.1007/s10059-012-0186-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 08/30/2012] [Accepted: 09/30/2012] [Indexed: 01/14/2023] Open
Abstract
Sonic hedgehog (Shh) signaling regulates patterning, proliferation, and stem cell self-renewal in many organs. Smoothened (Smo) plays a key role in transducing Shh signaling into the nucleus by activating a glioma family of transcription factors; however, the cellular and molecular mechanisms underlying the role of sustained Smo activation in postnatal development are still unclear. In this study, we explored the effects of Shh signaling on bone development using a conditional knock-in mouse model that expresses a constitutively activated form of Smo (SmoM2) upon osteocalcin (OCN)-Cre-mediated recombination (SmoM2; OCN-Cre mice). We also evaluated the expression pattern of bone formation-related factors in primary calvarial cultures of mutant and control mice. The SmoM2; OCN-Cre mutant showed growth retardation and reduction of bone mineral density compared to control mice. Constitutively activated SmoM2 also repressed mRNA expression of Runx2, osterix, type I collagen, and osteocalcin. Further, sustained SmoM2 induction suppressed mineralization in calvarial primary osteoblasts cultures, whereas such induction did not affect cell proliferation in the mutant cultures as compared with SmoM2 only control cultures. These results suggest that sustained Smo activation inhibits postnatal development of bone by suppressing gene expression of bone formation regulatory factors in mice.
Collapse
Affiliation(s)
- Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and Brain Korea 21 Program, Chonbuk National University,Jeonju 561-756,
Korea
| | - Shin-Saeng Lim
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and Brain Korea 21 Program, Chonbuk National University,Jeonju 561-756,
Korea
- Department of Bioactive Material Sciences, Chonbuk National University, Jeonju 561-756,
Korea
| | - Jae-Won Hwang
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and Brain Korea 21 Program, Chonbuk National University,Jeonju 561-756,
Korea
| | - Jeong-Chae Lee
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and Brain Korea 21 Program, Chonbuk National University,Jeonju 561-756,
Korea
- Department of Bioactive Material Sciences, Chonbuk National University, Jeonju 561-756,
Korea
| |
Collapse
|
13
|
Non-cell autonomous control of apoptosis by ligand-independent Hedgehog signaling in Drosophila. Cell Death Differ 2012; 20:302-11. [PMID: 23018595 DOI: 10.1038/cdd.2012.126] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hedgehog (Hh) signaling is important for development and homeostasis in vertebrates and invertebrates. Ligand-independent, deregulated Hh signaling caused by loss of negative regulators such as Patched causes excessive cell proliferation, leading to overgrowth in Drosophila and tumors in humans, including basal-cell carcinoma and medulloblastoma. We show that in Drosophila deregulated Hh signaling also promotes cell survival by increasing the resistance to apoptosis. Surprisingly, cells with deregulated Hh activity do not protect themselves from apoptosis; instead, they promote cell survival of neighboring wild-type cells. This non-cell autonomous effect is mediated by Hh-induced Notch signaling, which elevates the protein levels of Drosophila inhibitor of apoptosis protein-1 (Diap-1), conferring resistance to apoptosis. In summary, we demonstrate that deregulated Hh signaling not only promotes proliferation but also cell survival of neighboring cells. This non-cell autonomous control of apoptosis highlights an underappreciated function of deregulated Hh signaling, which may help to generate a supportive micro-environment for tumor development.
Collapse
|
14
|
Jing X, Grebenok RJ, Behmer ST. Plant sterols and host plant suitability for generalist and specialist caterpillars. JOURNAL OF INSECT PHYSIOLOGY 2012; 58:235-44. [PMID: 22154836 DOI: 10.1016/j.jinsphys.2011.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 11/15/2011] [Accepted: 11/17/2011] [Indexed: 05/16/2023]
Abstract
Insects, unlike plants and vertebrates, lack the ability to biosynthesize sterols. Cholesterol is typically the most common sterol found in plant-feeding insects, but it is rarely found in plants above trace levels, so plant-feeding insects must produce the cholesterol they need by metabolizing the sterols found in the plants they eat. Plant-feeding insects are, however, often limited in terms of which sterols can be converted to cholesterol. In the current study we used a transgenic tobacco plant line that displays high levels of atypical plant steroids, specifically stanols and ketone-steroids, to explore how novel steroid structural features affect performance in three economically important caterpillars (Heliothis virescens, Spodoptera exigua, and Manduca sexta). For each species we measured pupation success, larval development, pupal mass, pupal development, and eclosion success. For the two generalists species (H. virescens and S. exigua) we also measured egg production and egg viability. We then used these eggs to replicate the experiment, so that we could examine the effect of parental steroid dietary history on survival, growth and reproduction of 2nd-generation individuals. Significant negative effects of novel steroids on larval and pupal performance were observed for each caterpillar in the first generation, although these were often subtle, and were not consistent between the three species. In the second generation, larval survival estimated by 'pupation number/plant' on the tobacco plants with novel steroids was significantly reduced, while eclosion success was significantly lower for H. virescens. With respect to adult reproduction (i.e. egg production and egg viability) there were no observed differences in the first generation, but novel steroids significantly negatively impacted reproduction in the second generation. The findings from this study, when integrated into a simple population growth model, demonstrate the potential in using plants with modified steroids as a novel approach to manage populations of economically important caterpillar species.
Collapse
Affiliation(s)
- Xiangfeng Jing
- Department of Entomology, Texas A&M University, College Station, TX 77843-2475, USA.
| | | | | |
Collapse
|
15
|
Soleti R, Martinez MC. Sonic Hedgehog on microparticles and neovascularization. VITAMINS AND HORMONES 2012; 88:395-438. [PMID: 22391314 DOI: 10.1016/b978-0-12-394622-5.00018-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neovascularization represents a pivotal process consisting in the development of vascular network during embryogenesis and adult life. Postnatally, it arises mainly through angiogenesis, which has physiological and pathological roles in health and disease. Blood vessel formation results as tightly regulated multistep process which needs coordination and precise regulation of the balance of proangiogenic and antiangiogenic factors. Sonic Hedgehog (SHH), a morphogen belonging to Hedgehog (HH) family proteins, is implicated in a remarkably wide variety of process, including vessel development. Recent evidence demonstrate that, in addition to the classic factors, microvesicles (MVs), both microparticles (MPs) and exosomes, small vesicles released distinct cellular compartments, are involved in modulation of neovascularization. MPs generated from T lymphocytes undergoing both activation and apoptosis harbor at their surface SHH and play a crucial role in modulation of neovascularization. They are able to modulate the different steps implicated in angiogenesis process in vitro and to enhance postischemic neovascularization in vivo. As the consequence, we suggest that the MPs carrying SHH contribute to generation of a vascular network and may represent a new therapeutic approach to treat pathologies associated with failed angiogenesis.
Collapse
|
16
|
Chang SC, Mulloy B, Magee AI, Couchman JR. Two distinct sites in sonic Hedgehog combine for heparan sulfate interactions and cell signaling functions. J Biol Chem 2011; 286:44391-402. [PMID: 22049079 PMCID: PMC3247953 DOI: 10.1074/jbc.m111.285361] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 10/10/2011] [Indexed: 01/01/2023] Open
Abstract
Hedgehog (Hh) proteins are morphogens that mediate many developmental processes. Hh signaling is significant for many aspects of embryonic development, whereas dysregulation of this pathway is associated with several types of cancer. Hh proteins require heparan sulfate proteoglycans (HSPGs) for their normal distribution and signaling activity. Here, we have used molecular modeling to examine the heparin-binding domain of sonic hedgehog (Shh). In biochemical and cell biological assays, the importance of specific residues of the putative heparin-binding domain for signaling was assessed. It was determined that key residues in human (h) Shh involved in heparin and HSPG syndecan-4 binding and biological activity included the well known cationic Cardin-Weintraub motif (lysines 32-38) but also a previously unidentified major role for lysine 178. The activity of Shh mutated in these residues was tested by quantitation of alkaline phosphatase activity in C3H10T1/2 cells differentiating into osteoblasts and hShh-inducible gene expression in PANC1 human pancreatic ductal adenocarcinoma cells. Mutated hShhs such as K37S/K38S, K178S, and particularly K37S/K38S/K178S that could not interact with heparin efficiently had reduced signaling activity compared with wild type hShh or a control mutation (K74S). In addition, the mutant hShh proteins supported reduced proliferation and invasion of PANC1 cells compared with control hShh proteins, following endogenous hShh depletion by RNAi knockdown. The data correlated with reduced Shh multimerization where the Lys-37/38 and/or Lys-178 mutations were examined. These studies provide a new insight into the functional roles of hShh interactions with HSPGs, which may allow targeting this aspect of hShh biology in, for example, pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Shu-Chun Chang
- From the Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Barbara Mulloy
- the National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom, and
| | - Anthony I. Magee
- From the Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - John R. Couchman
- the Department of Biomedical Sciences, University of Copenhagen, Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| |
Collapse
|
17
|
Choy W, Kim W, Nagasawa D, Stramotas S, Yew A, Gopen Q, Parsa AT, Yang I. The molecular genetics and tumor pathogenesis of meningiomas and the future directions of meningioma treatments. Neurosurg Focus 2011; 30:E6. [DOI: 10.3171/2011.2.focus1116] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Meningiomas are mostly benign, slow-growing tumors of the CNS that originate from arachnoidal cap cells. While monosomy 22 is the most frequent genetic abnormality found in meningiomas, a multitude of other aberrant chromosomal alterations, signaling pathways, and growth factors have been implicated in its pathogenesis. Losses on 22q12.2, a region encoding the tumor suppressor gene merlin, represent the most common genetic alterations in early meningioma formation. Malignant meningioma progression, however, is associated with more complex karyotypes and greater genetic instability. Cytogenetic studies of atypical and anaplastic meningiomas revealed gains and losses on chromosomes 9, 10, 14, and 18, with amplifications on chromosome 17. However, the specific gene targets in a majority of these chromosomal abnormalities remain elusive.
Studies have also implicated a myriad of aberrant signaling pathways involved with meningioma tumorigenesis, including those involved with proliferation, angiogenesis, and autocrine loops. Understanding these disrupted pathways will aid in deciphering the relationship between various genetic changes and their downstream effects on meningioma pathogenesis.
Despite advancements in our understanding of meningioma pathogenesis, the conventional treatments, including surgery, radiotherapy, and stereotactic radiosurgery, have remained largely stagnant. Surgery and radiation therapy are curative in the majority of lesions, yet treatment remains challenging for meningiomas that are recurrent, aggressive, or refractory to conventional treatments. Future therapies will include combinations of targeted molecular agents as a result of continued progress in the understanding of genetic and biological changes associated with meningiomas.
Collapse
Affiliation(s)
| | - Won Kim
- 1Department of Neurological Surgery, and
| | | | | | - Andrew Yew
- 1Department of Neurological Surgery, and
| | - Quinton Gopen
- 2Division of Otolaryngology, University of California Los Angeles; and
| | - Andrew T. Parsa
- 3Department of Neurological Surgery, University of California, San Francisco, California
| | - Isaac Yang
- 1Department of Neurological Surgery, and
| |
Collapse
|
18
|
A novel signaling pathway mediated by the nuclear targeting of C-terminal fragments of mammalian Patched 1. PLoS One 2011; 6:e18638. [PMID: 21533246 PMCID: PMC3076429 DOI: 10.1371/journal.pone.0018638] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 03/14/2011] [Indexed: 12/20/2022] Open
Abstract
Background Patched 1 (Ptc1) is a polytopic receptor protein that is essential for growth and differentiation. Its extracellular domains accept its ligand, Sonic Hedgehog, while the function of its C-terminal intracellular domain is largely obscure. Principal Findings In this study, we stably expressed human Ptc1 protein in HeLa cells and found that it is subjected to proteolytic cleavage at the C-terminus, resulting in the generation of soluble C-terminal fragments. These fragments accumulated in the nucleus, while the N-terminal region of Ptc1 remained in the cytoplasmic membrane fractions. Using an anti-Ptc1 C-terminal domain antibody, we provide conclusive evidence that C-terminal fragments of endogenous Ptc1 accumulate in the nucleus of C3H10T1/2 cells. Similar nuclear accumulation of endogenous C-terminal fragments was observed not only in C3H10T1/2 cells but also in mouse embryonic primary cells. Importantly, the C-terminal fragments of Ptc1 modulate transcriptional activity of Gli1. Conclusions Although Ptc1 protein was originally thought to be restricted to cell membrane fractions, our findings suggest that its C-terminal fragments can function as an alternative signal transducer that is directly transported to the cell nucleus.
Collapse
|
19
|
Chia YH, Ma CX. Hedgehog Pathway Inhibitors: Potential Applications in Breast Cancer. CURRENT BREAST CANCER REPORTS 2010. [DOI: 10.1007/s12609-010-0031-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
20
|
Mansour AA, Nissim-Eliraz E, Zisman S, Golan-Lev T, Schatz O, Klar A, Ben-Arie N. Foxa2 regulates the expression of Nato3 in the floor plate by a novel evolutionarily conserved promoter. Mol Cell Neurosci 2010; 46:187-99. [PMID: 20849957 DOI: 10.1016/j.mcn.2010.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 08/29/2010] [Accepted: 09/01/2010] [Indexed: 11/24/2022] Open
Abstract
The development of the neural tube into a complex central nervous system involves morphological, cellular and molecular changes, all of which are tightly regulated. The floor plate (FP) is a critical organizing center located at the ventral-most midline of the neural tube. FP cells regulate dorsoventral patterning, differentiation and axon guidance by secreting morphogens. Here we show that the bHLH transcription factor Nato3 (Ferd3l) is specifically expressed in the spinal FP of chick and mouse embryos. Using in ovo electroporation to understand the regulation of the FP-specific expression of Nato3, we have identified an evolutionarily conserved 204 bp genomic region, which is necessary and sufficient to drive expression to the chick FP. This promoter contains two Foxa2-binding sites, which are highly conserved among distant phyla. The two sites can bind Foxa2 in vitro, and are necessary for the expression in the FP in vivo. Gain and loss of Foxa2 function in vivo further emphasize its role in Nato3 promoter activity. Thus, our data suggest that Nato3 is a direct target of Foxa2, a transcription activator and effector of Sonic hedgehog, the hallmark regulator of FP induction and spinal cord development. The identification of the FP-specific promoter is an important step towards a better understanding of the molecular mechanisms through which Nato3 transcription is regulated and for uncovering its function during nervous system development. Moreover, the promoter provides us with a powerful tool for conditional genetic manipulations in the FP.
Collapse
Affiliation(s)
- Abed AlFatah Mansour
- Department of Cell and Developmental Biology, Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | | | | | | | | | | | | |
Collapse
|
21
|
Queiroz KCS, Tio RA, Zeebregts CJ, Bijlsma MF, Zijlstra F, Badlou B, de Vries M, Ferreira CV, Spek CA, Peppelenbosch MP, Rezaee F. Human Plasma Very Low Density Lipoprotein Carries Indian Hedgehog. J Proteome Res 2010; 9:6052-9. [DOI: 10.1021/pr100403q] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Karla C. S. Queiroz
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - René A. Tio
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - Clark J. Zeebregts
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - Maarten F. Bijlsma
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - Felix Zijlstra
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - Bahram Badlou
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - Marcel de Vries
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - Carmen V. Ferreira
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - C. Arnold Spek
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - Maikel P. Peppelenbosch
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| | - Farhad Rezaee
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands, Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands, Department of Surgery, Division of Vascular Surgery, University Medical Center Groningen, Groningen, The Netherlands, Department of Biochemistry, Institute of Biology, State University of Campinas, Brazil, Campinas, SÄo Paulo, Brazil, Center
| |
Collapse
|
22
|
Lipinski RJ, Bushman W. Identification of Hedgehog signaling inhibitors with relevant human exposure by small molecule screening. Toxicol In Vitro 2010; 24:1404-9. [PMID: 20434536 PMCID: PMC2891024 DOI: 10.1016/j.tiv.2010.04.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 02/25/2010] [Accepted: 04/24/2010] [Indexed: 01/10/2023]
Abstract
In animal models, chemical disruption of the Hedgehog (Hh) signaling pathway during embryonic development causes severe birth defects including holoprosencephaly and cleft lip and palate. The exact etiological basis of correlate human birth defects remains uncertain but is likely multifactorial, involving the interaction of genetic and environmental or chemical influences. The Hh transduction mechanism relies upon endogenous small molecule regulation, conferring remarkable pathway sensitivity to inhibition by a structurally diverse set of exogenous small molecules. Here, we employed small molecule screening to identify human exposure-relevant Hh signaling inhibitors. From a library of 4240 compounds, including pharmaceuticals, natural products, and pesticides, three putative Hh pathway inhibitors were identified: tolnaftate, an antifungal agent; ipriflavone, a dietary supplement; and 17-beta-estradiol, a human hormone and pharmaceutical agent. Each compound inhibited Hh signaling in both mouse and human cells. Dose-response assays determined the three compounds to be 8- to 30-fold less potent than the index Hh pathway inhibitor cyclopamine. Despite current limitations in chemical library availability, which narrowed the scope of this study to only a small fraction of all human exposure-relevant small molecules, three structurally diverse environmental Hh signaling inhibitors were identified, highlighting an inherent pathway vulnerability to teratogenic influences.
Collapse
Affiliation(s)
- Robert J. Lipinski
- Molecular and Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Urology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI,USA
| | - Wade Bushman
- Molecular and Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Urology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI,USA
| |
Collapse
|
23
|
Laurendeau I, Ferrer M, Garrido D, D'Haene N, Ciavarelli P, Basso A, Vidaud M, Bieche I, Salmon I, Szijan I. Gene expression profiling of the hedgehog signaling pathway in human meningiomas. Mol Med 2010; 16:262-70. [PMID: 20386868 DOI: 10.2119/molmed.2010.00005] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 03/25/2010] [Indexed: 12/22/2022] Open
Abstract
The Hedgehog (Hh) signaling pathway has an important role during embryogenesis and in adult life, regulating proliferation, angiogenesis, matrix remodeling and stem-cell renewal. Deregulation of the Hh pathway is involved in tumor development, since mutations in several components of this pathway were found in patients with basal cell carcinoma, medulloblastoma and other tumors; however, the role of Hh in meningiomas has not been studied yet. Meningiomas represent 30% of primary cranial tumors, are mostly benign and prevail in the second half of life. Novel therapies for meningiomas such as targeted molecular agents could use Hh pathway components. To provide information concerning molecular alterations, by use of real-time RT-PCR, we studied expression at the mRNA level of 32 Hh pathway and target genes in 36 meningioma specimens of different grades. mRNA levels of 16 genes, involved mainly in Hh pathway activation and cell proliferation, increased in meningiomas in comparison with normal tissue, whereas those of 7 genes, mainly related to Hh pathway repression, decreased. The most significant changes occurred in signal transduction (SMO) and GLI-transcription factor genes, and the target FOXM1 mRNA attained the highest values; their over-expression was found in aggressive and in benign tumors. Some proliferation-related genes (SPP1, IGF2) were overexpressed in higher meningioma grades. A correlation in expression between genes with a similar function was also found. Our results show a marked activation of the Hh pathway in meningiomas, which may be important for their biological and clinical characterization and would be useful for gene therapy.
Collapse
Affiliation(s)
- Ingrid Laurendeau
- UMR745, INSERM, Université Paris-Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kameda C, Nakamura M, Tanaka H, Yamasaki A, Kubo M, Tanaka M, Onishi H, Katano M. Oestrogen receptor-alpha contributes to the regulation of the hedgehog signalling pathway in ERalpha-positive gastric cancer. Br J Cancer 2010; 102:738-47. [PMID: 20087349 PMCID: PMC2837575 DOI: 10.1038/sj.bjc.6605517] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Oestrogen receptor-alpha (ERalpha) is highly expressed in diffuse-type gastric cancer and oestrogen increases the proliferation of ERalpha-positive gastric cancer. However, a detailed mechanism by which oestrogen increases the proliferation of these cells is still unclear. METHODS We used 17-beta-oestradiol (E2) as a stimulator against the ERalpha pathway. Pure anti-oestrogen drug ICI 182 780 (ICI) and small interfering RNA against ERalpha (ERalpha siRNA) were used as inhibitors. Cyclopamine (Cyc) was used as the hedgehog (Hh) pathway inhibitor. Two human ERalpha-positive gastric cancer cells were used as target cells. Effects of the stimulator and inhibitor on E2-induced cell proliferation were also examined. RESULTS In ERalpha-positive cells, E2 increased not only cell proliferation but also one of the ligands of the Hh pathway, Shh expression. 17-beta-Oestradiol-induced cell proliferation was suppressed by ICI, ERalpha siRNA or Cyc. The increased expression of Shh induced by E2 was suppressed by ICI and ERalpha siRNA but not by Cyc. Furthermore, recombinant Shh activated the Hh pathway and increased cell proliferation, whereas anti-Shh antibody suppressed E2-induced cell proliferation. When a relationship between ERalpha and Shh expressions was analysed using surgically resected gastric cancer specimens, a positive correlation was found, suggesting a linkage between the ERalpha and Hh pathways. CONCLUSION Our data indicate that activation of the ERalpha pathway promotes cell proliferation by activating the Hh pathway in a ligand-dependent manner through Shh induction of ERalpha-positive gastric cancer.
Collapse
Affiliation(s)
- C Kameda
- Department of Cancer Therapy and Research, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Mahindroo N, Connelly MC, Punchihewa C, Kimura H, Smeltzer MP, Wu S, Fujii N. Structure-activity relationships and cancer-cell selective toxicity of novel inhibitors of glioma-associated oncogene homologue 1 (Gli1) mediated transcription. J Med Chem 2009; 52:4277-87. [PMID: 19545120 PMCID: PMC2853048 DOI: 10.1021/jm900106f] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We report novel inhibitors of Gli1-mediated transcription as potential anticancer agents. Focused chemical libraries were designed and assessed for inhibition of functional cell-based Gli1-mediated transcription and selective toxicity toward cancer cells. The SAR was revealed, and the selectivity of the lead compounds' inhibition of Gli1-mediated transcription over that of Gli2 was determined. Compound 63 (NMDA298-1), which inhibited Gli1-mediated transcription in C3H10T1/2 cells with an IC(50) of 6.9 muM, showed 3-fold selectivity for inhibiting transcription mediated by Gli1 over that by Gli2. Cell-viability assays were performed to evaluate the chemical library in a normal cell line and a panel of cancer cell lines with or without up-regulated expression of the Gli1 gene. These compounds decreased the viability of several cancer cell lines but were less active in the noncancerous BJ-hTERT cells.
Collapse
Affiliation(s)
- Neeraj Mahindroo
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Michele C. Connelly
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Chandanamali Punchihewa
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Hiromichi Kimura
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Matthew P. Smeltzer
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Song Wu
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Naoaki Fujii
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
26
|
Zhang X, Harrington N, Moraes RC, Wu MF, Hilsenbeck SG, Lewis MT. Cyclopamine inhibition of human breast cancer cell growth independent of Smoothened (Smo). Breast Cancer Res Treat 2009; 115:505-21. [PMID: 18563554 PMCID: PMC5300001 DOI: 10.1007/s10549-008-0093-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 06/04/2008] [Indexed: 11/25/2022]
Abstract
Altered hedgehog signaling is implicated in the development of approximately 20-25% of all cancers, especially those of soft tissues. Genetic evidence in mice as well as immunolocalization studies in human breast cancer specimens suggest that deregulated hedgehog signaling may contribute to breast cancer development. Indeed, two recent studies demonstrated that anchorage-dependent growth of some human breast cancer cell lines is impaired by cyclopamine, a potent hedgehog signaling antagonist targeting the Smoothened (SMO) protein. However, specificity of cyclopamine at the dosage required for growth inhibition (> or =10 microM) remained an open question. In this paper we demonstrate that hedgehog signaling antagonists, including cyclopamine, and a second compound, CUR0199691, can inhibit growth of estrogen receptor (ER)-positive and ER-negative tumorigenic breast cancer cells at elevated doses. However, our results indicate that, for most breast cancer cell lines, growth inhibition by these compounds can be independent of detectable Smo gene expression. Rather, our results suggest that cyclopamine and CUR0199691 have unique secondary molecular targets at the dosages required for growth inhibition that are unrelated to hedgehog signaling.
Collapse
MESH Headings
- Apoptosis/drug effects
- Binding, Competitive
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Adhesion/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cholesterol/metabolism
- Drug Resistance, Neoplasm/drug effects
- Female
- Flow Cytometry
- Fluorescent Antibody Technique
- Gene Expression Regulation, Neoplastic/drug effects
- Hedgehog Proteins/genetics
- Hedgehog Proteins/metabolism
- Humans
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Estrogen/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Smoothened Receptor
- Veratrum Alkaloids/pharmacology
Collapse
Affiliation(s)
- Xiaomei Zhang
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Nikesha Harrington
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Ricardo C. Moraes
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Meng-Fen Wu
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Susan G. Hilsenbeck
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center and the Department of Molecular and Cellular Biology, Room N1210; BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, TEL: (713)798-3296, FAX: (713)798-1673,
| |
Collapse
|
27
|
|
28
|
Prykhozhij SV, Neumann CJ. Distinct roles of Shh and Fgf signaling in regulating cell proliferation during zebrafish pectoral fin development. BMC DEVELOPMENTAL BIOLOGY 2008; 8:91. [PMID: 18811955 PMCID: PMC2562996 DOI: 10.1186/1471-213x-8-91] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Accepted: 09/23/2008] [Indexed: 11/13/2022]
Abstract
Background Cell proliferation in multicellular organisms must be coordinated with pattern formation. The major signaling pathways directing pattern formation in the vertebrate limb are well characterized, and we have therefore chosen this organ to examine the interaction between proliferation and patterning. Two important signals for limb development are members of the Hedgehog (Hh) and Fibroblast Growth Factor (Fgf) families of secreted signaling proteins. Sonic hedgehog (Shh) directs pattern formation along the anterior/posterior axis of the limb, whereas several Fgfs in combination direct pattern formation along the proximal/distal axis of the limb. Results We used the genetic and pharmacological amenability of the zebrafish model system to dissect the relative importance of Shh and Fgf signaling in regulating proliferation during development of the pectoral fin buds. In zebrafish mutants disrupting the shh gene, proliferation in the pectoral fin buds is initially normal, but later is strongly reduced. Correlating with this reduction, Fgf signaling is normal at early stages, but is later lost in shh mutants. Furthermore, pharmacological inhibition of Hh signaling for short periods has little effect on either Fgf signaling, or on expression of G1- and S-phase cell-cycle genes, whereas long periods of inhibition lead to the downregulation of both. In contrast, even short periods of pharmacological inhibition of Fgf signaling lead to strong disruption of proliferation in the fin buds, without affecting Shh signaling. To directly test the ability of Fgf signaling to regulate proliferation in the absence of Shh signaling, we implanted beads soaked with Fgf protein into shh mutant fin buds. We find that Fgf-soaked beads rescue proliferation in the pectoral find buds of shh mutants, indicating that Fgf signaling is sufficient to direct proliferation in zebrafish fin buds in the absence of Shh. Conclusion Previous studies have shown that both Shh and Fgf signaling are crucial for outgrowth of the vertebrate limb. The results presented here show that the role of Shh in this process is indirect, and is mediated by its effect on Fgf signaling. By contrast, the activity of the Fgf pathway affects proliferation directly and independently of its effect on Shh. These results show that Fgf signaling is of primary importance in directing outgrowth of the limb bud, and clarify the role of the Shh-Fgf feedback loop in regulating proliferation.
Collapse
Affiliation(s)
- Sergey V Prykhozhij
- Developmental Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, Heidelberg, Germany.
| | | |
Collapse
|
29
|
Dentice M, Monfrecola G. Dual dichotomies--when thyroid dysfunction and thyroid hormones get into the skin. Thyroid 2008; 18:823-4. [PMID: 18690795 DOI: 10.1089/thy.2008.1542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
30
|
Franco PG, Silvestroff L, Soto EF, Pasquini JM. Thyroid hormones promote differentiation of oligodendrocyte progenitor cells and improve remyelination after cuprizone-induced demyelination. Exp Neurol 2008; 212:458-67. [PMID: 18572165 DOI: 10.1016/j.expneurol.2008.04.039] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 04/16/2008] [Accepted: 04/24/2008] [Indexed: 11/26/2022]
Abstract
In the present work we analyzed the capacity of thyroid hormones (THs) to improve remyelination using a rat model of cuprizone-induced demyelination previously described in our laboratories. Twenty one days old Wistar rats were fed a diet containing 0.6% cuprizone for two weeks to induce demyelination. After cuprizone withdrawal, rats were injected with triiodothyronine (T3). Histological studies carried out in these animals revealed that remyelination in the corpus callosum (CC) of T3-treated rats improved markedly when compared to saline treated animals. The cellular events occurring in the CC and in the subventricular zone (SVZ) during the first week of remyelination were analyzed using specific oligodendroglial cell (OLGc) markers. In the CC of saline treated demyelinated animals, mature OLGcs decreased and oligodendroglial precursor cells (OPCs) increased after one week of spontaneous remyelination. Furthermore, the SVZ of these animals showed an increase in early progenitor cell numbers, dispersion of OPCs and inhibition of Olig and Shh expression compared to non-demyelinated animals. The changes triggered by demyelination were reverted after T3 administration, suggesting that THs could be regulating the emergence of remyelinating oligodendrocytes from the pool of proliferating cells residing in the SVZ. Our results also suggest that THs receptor beta mediates T3 effects on remyelination. These results support a potential role for THs in the remyelination process that could be used to develop new therapeutic approaches for demyelinating diseases.
Collapse
Affiliation(s)
- P G Franco
- Departamento de Química Biológica, IQUIFIB and IIMHNO, Facultad de Farmacia y Bioquímica, UBA-CONICET, Junín 956, Buenos Aires C1113AAD, Argentina
| | | | | | | |
Collapse
|
31
|
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
|
32
|
Li Z, Zhang H, Denhard LA, Liu LH, Zhou H, Lan ZJ. Reduced white fat mass in adult mice bearing a truncated Patched 1. Int J Biol Sci 2008; 4:29-36. [PMID: 18274621 PMCID: PMC2238183 DOI: 10.7150/ijbs.4.29] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Accepted: 01/25/2008] [Indexed: 01/25/2023] Open
Abstract
Hedgehog (Hh) signaling emerges as a potential pathway contributing to fat formation during postnatal development. In this report, we found that Patched 1 (Ptc1), a negative regulator of Hh signaling, was expressed in the epididymal fat pad of adult mice. Reduced total white fat mass and epididymal adipocyte cell size were observed in naturally occurring spontaneous mesenchymal dysplasia (mes) adult mice (Ptc1mes/mes), which carry a deletion of Ptc1 at the carboxyl-terminal cytoplasmic region. Increased expression of truncated Ptc1, Ptc2 and Gli1, the indicators of ectopic activation of Hh signaling, was observed in epididymal fat pads of adult Ptc1mes/mes mice. In contrast, expression of peroxisome proliferator-activated receptor gamma, CCAAT/enhancer binding protein alpha, adipocyte P2 and adipsin were reduced in epididymal fat pads of adult Ptc1mes/mes mice. Taken together, our results indicate that deletion of carboxyl-terminal tail of Ptc1 can lead to the reduction of white fat mass during postnatal development.
Collapse
Affiliation(s)
- Zili Li
- Birth Defects Center, Department of Molecular, Cellular, Craniofacial Biology, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | | | | | | | | | | |
Collapse
|
33
|
|
34
|
Dentice M, Luongo C, Huang S, Ambrosio R, Elefante A, Mirebeau-Prunier D, Zavacki AM, Fenzi G, Grachtchouk M, Hutchin M, Dlugosz AA, Bianco AC, Missero C, Larsen PR, Salvatore D. Sonic hedgehog-induced type 3 deiodinase blocks thyroid hormone action enhancing proliferation of normal and malignant keratinocytes. Proc Natl Acad Sci U S A 2007; 104:14466-71. [PMID: 17720805 PMCID: PMC1964817 DOI: 10.1073/pnas.0706754104] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2006] [Indexed: 12/21/2022] Open
Abstract
The Sonic hedgehog (Shh) pathway plays a critical role in hair follicle physiology and is constitutively active in basal cell carcinomas (BCCs), the most common human malignancy. Type 3 iodothyronine deiodinase (D3), the thyroid hormone-inactivating enzyme, is frequently expressed in proliferating and neoplastic cells, but its role in this context is unknown. Here we show that Shh, through Gli2, directly induces D3 in proliferating keratinocytes and in mouse and human BCCs. We demonstrate that Gli-induced D3 reduces intracellular active thyroid hormone, thus resulting in increased cyclin D1 and keratinocyte proliferation. D3 knockdown caused a 5-fold reduction in the growth of BCC xenografts in nude mice. Shh-induced thyroid hormone degradation via D3 synergizes with the Shh-mediated reduction of the type 2 deiodinase, the thyroxine-activating enzyme, and both effects are reversed by cAMP. This previously unrecognized functional cross-talk between Shh/Gli2 and thyroid hormone in keratinocytes is a pathway by which Shh produces its proliferative effects and offers a potential therapeutic approach to BCC.
Collapse
Affiliation(s)
- Monica Dentice
- *Department of Molecular and Clinical Endocrinology and Oncology, University of Naples Federico II, 80131 Naples, Italy
| | - Cristina Luongo
- *Department of Molecular and Clinical Endocrinology and Oncology, University of Naples Federico II, 80131 Naples, Italy
| | - Stephen Huang
- Thyroid Section, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Raffaele Ambrosio
- *Department of Molecular and Clinical Endocrinology and Oncology, University of Naples Federico II, 80131 Naples, Italy
| | - Antonia Elefante
- *Department of Molecular and Clinical Endocrinology and Oncology, University of Naples Federico II, 80131 Naples, Italy
| | - Delphine Mirebeau-Prunier
- Thyroid Section, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Ann Marie Zavacki
- Thyroid Section, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Gianfranco Fenzi
- *Department of Molecular and Clinical Endocrinology and Oncology, University of Naples Federico II, 80131 Naples, Italy
| | - Marina Grachtchouk
- Department of Dermatology and Comprehensive Cancer Center, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109; and
| | - Mark Hutchin
- Department of Dermatology and Comprehensive Cancer Center, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109; and
| | - Andrzej A. Dlugosz
- Department of Dermatology and Comprehensive Cancer Center, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109; and
| | - Antonio C. Bianco
- Thyroid Section, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Caterina Missero
- Center of Genetics Engineering (CEINGE), Biotecnologie Avanzate Scarl, 80145 Naples, Italy
| | - P. Reed Larsen
- Thyroid Section, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Domenico Salvatore
- *Department of Molecular and Clinical Endocrinology and Oncology, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
35
|
Cousin W, Fontaine C, Dani C, Peraldi P. Hedgehog and adipogenesis: fat and fiction. Biochimie 2007; 89:1447-53. [PMID: 17933451 DOI: 10.1016/j.biochi.2007.08.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Accepted: 08/24/2007] [Indexed: 11/19/2022]
Abstract
Morphogenes, abundantly described during embryogenesis have recently emerged as crucial modulators of cell differentiation processes. Hedgehog signaling, the dysregulation of which causing several pathologies such as congenital defects and cancer, is involved in several cell differentiation processes including adipogenesis. This review presents an overview of the relations between Hedgehog signaling, adipocyte differentiation and fat mass. While the anti-adipogenic role of Hedgehog signaling seems to be established, the effect of Hedgehog inhibition on adipocyte differentiation in vitro remains debated. Finally, Hedgehog potential as a pharmacological target to treat fat mass disorders is discussed.
Collapse
Affiliation(s)
- Wendy Cousin
- ISBDC, Université De Nice Sophia-Antipolis, CNRS, 28 Avenue De Valrose, 06100 Nice, France.
| | | | | | | |
Collapse
|
36
|
Gerber AN, Wilson CW, Li YJ, Chuang PT. The hedgehog regulated oncogenes Gli1 and Gli2 block myoblast differentiation by inhibiting MyoD-mediated transcriptional activation. Oncogene 2007; 26:1122-36. [PMID: 16964293 PMCID: PMC3325095 DOI: 10.1038/sj.onc.1209891] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Revised: 07/03/2006] [Accepted: 07/05/2006] [Indexed: 01/21/2023]
Abstract
The mechanism by which activation of the Hedgehog (Hh) pathway modulates differentiation and promotes oncogenesis in specific tissues is poorly understood. We therefore, analysed rhabdomyosarcomas from mice that were haploinsufficient for the Hh-binding protein, Hip1, or for the Hh receptor, Patched 1 (Ptch1). Transfection of the Hh-regulated transcription factor Gli1, which is expressed in a subset of mouse and human rhabdomyosarcomas, suppressed differentiation of myogenic rhabdomyosarcoma lines generated from Hip1+/- and Ptch1+/- mice. The closely related factor, Gli2, had similar effects. Gli1 and Gli2 inhibited myogenesis by repressing the capacity of MyoD to activate transcription. Deletion analysis of Gli1 indicated that multiple domains of Gli1 are required for efficient inhibition of MyoD. Gli1 reduced the ability of MyoD to heterodimerize with E12 and bind DNA, providing one mechanism whereby the Gli proteins modulate the activity of MyoD. This novel activity of Gli proteins provides new insights into how Hh signaling modulates terminal differentiation through inhibition of tissue-specific factors such as MyoD. This mechanism may contribute to the broad role of Hh signaling and the Gli proteins in differentiation decisions and cancer formation.
Collapse
Affiliation(s)
- AN Gerber
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - CW Wilson
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Y-J Li
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - P-T Chuang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
37
|
Moraes RC, Zhang X, Harrington N, Fung JY, Wu MF, Hilsenbeck SG, Allred DC, Lewis MT. Constitutive activation of smoothened (SMO) in mammary glands of transgenic mice leads to increased proliferation, altered differentiation and ductal dysplasia. Development 2007; 134:1231-42. [PMID: 17287253 DOI: 10.1242/dev.02797] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The hedgehog signaling network regulates pattern formation, proliferation, cell fate and stem/progenitor cell self-renewal in many organs. Altered hedgehog signaling is implicated in 20-25% of all cancers, including breast cancer. We demonstrated previously that heterozygous disruption of the gene encoding the patched-1 (PTCH1) hedgehog receptor, a negative regulator of smoothened (Smo) in the absence of ligand, led to mammary ductal dysplasia in virgin mice. We now show that expression of activated human SMO (SmoM2) under the mouse mammary tumor virus (MMTV) promoter in transgenic mice leads to increased proliferation, altered differentiation, and ductal dysplasias distinct from those caused by Ptch1 heterozygosity. SMO activation also increased the mammosphere-forming efficiency of primary mammary epithelial cells. However, limiting-dilution transplantation showed a decrease in the frequency of regenerative stem cells in MMTV-SmoM2 epithelium relative to wild type, suggesting enhanced mammosphere-forming efficiency was due to increased survival or activity of division-competent cell types under anchorage-independent growth conditions, rather than an increase in the proportion of regenerative stem cells per se. In human clinical samples, altered hedgehog signaling occurs early in breast cancer development, with PTCH1 expression reduced in approximately 50% of ductal carcinoma in situ (DCIS) and invasive breast cancers (IBC). Conversely, SMO is ectopically expressed in 70% of DCIS and 30% of IBC. Surprisingly, in both human tumors and MMTV-SmoM2 mice, SMO rarely colocalized with the Ki67 proliferation marker. Our data suggest that altered hedgehog signaling may contribute to breast cancer development by stimulating proliferation, and by increasing the pool of division-competent cells capable of anchorage-independent growth.
Collapse
MESH Headings
- Animals
- Apoptosis
- Caspase 3/metabolism
- Cell Differentiation
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Fibrocystic Breast Disease/etiology
- Fibrocystic Breast Disease/genetics
- Fibrocystic Breast Disease/pathology
- Hedgehog Proteins/metabolism
- Humans
- Ki-67 Antigen/analysis
- Ki-67 Antigen/metabolism
- Mammary Glands, Animal/chemistry
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Transgenic
- Patched Receptors
- Patched-1 Receptor
- Promoter Regions, Genetic
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, G-Protein-Coupled/analysis
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Smoothened Receptor
- Transgenes
Collapse
Affiliation(s)
- Ricardo C Moraes
- Baylor Breast Center and Department of Molecular and Cellular Biology, Room N1210; MS:BCM600, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Lewis MT, Visbal AP. The hedgehog signaling network, mammary stem cells, and breast cancer: connections and controversies. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2007:181-217. [PMID: 17939302 DOI: 10.1007/2789_2007_051] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Several signal transduction networks have been implicated in the regulation of mammary epithelial stem cell self-renewal and maintenance (Kalirai and Clarke 2006; Liu et al. 2005). These signaling networks include those of the Wnt, Notch, TGFO, EGF, FGF, IGF, and most recently, the Hedgehog (Hh) families of secreted ligands. However, we currently know very little about the cellular and molecular mechanisms by which these signaling pathways function to regulate normal epithelial stem/progenitor cells. What is clear is that the regulatory signaling networks thought to control normal stem/progenitor cell self-renewal and maintenance are, with the current sole exception of the hedgehog network, well-documented to have contributory roles in mammary cancer development and disease progression when misregulated. In this review, genetic regulation of mammary gland development by hedgehog network genes is outlined, highlighting a developing controversy as to whether activated hedgehog signaling regulates normal regenerative mammary epithelial stem cells or, indeed, whether activated hedgehog signaling functions at all in ductal development. In addition, the question of whether inappropriate hedgehog network activation influences breast cancer development is addressed, with emphasis on the prospects for using hedgehog signaling antagonists clinically for breast cancer treatment or prevention.
Collapse
Affiliation(s)
- M T Lewis
- Baylor Breast Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Room N1210; MS:BCM600, One Baylor Plaza, 77030 Houston, TX, USA.
| | | |
Collapse
|
39
|
Abstract
In this review, we focus on the basic biology of the important developmental Hedgehog (Hh) protein family, its general function in development, pathway mechanisms, and gene discovery and nomenclature. Hh function in cardiovascular development and recent findings concerning Hh signaling in ischemia models are discussed in more detail, and future perspectives are proposed. In light of the recent discovery of Hh transport by insect lipophorin, we also hypothesize a role for low-density lipoprotein (LDL) in mammalian Hh transport, creating a surprising role for LDL in cardiovascular disease.
Collapse
Affiliation(s)
- Maarten F Bijlsma
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
40
|
Cousin W, Dani C, Peraldi P. Inhibition of the anti-adipogenic Hedgehog signaling pathway by cyclopamine does not trigger adipocyte differentiation. Biochem Biophys Res Commun 2006; 349:799-803. [PMID: 16949046 DOI: 10.1016/j.bbrc.2006.08.112] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Accepted: 08/17/2006] [Indexed: 01/24/2023]
Abstract
Dysregulation of Hedgehog signaling can lead to several pathologies such as congenital defects and cancer. Here, we show that Hedgehog signaling is active in undifferentiated 3T3-L1 cells and decreases during adipocyte differentiation. Interestingly, this is paralleled by a decrease in Indian Hedgehog expression. We then tested if this down-regulation was sufficient to induce adipocyte differentiation. To this end, we demonstrate that the well-characterized Hedgehog inhibitor cyclopamine induced a decrease in Hedgehog signaling, similar to the one observed during adipocyte differentiation. However, cyclopamine did not induce nor potentiate adipocyte differentiation, as monitored by triglyceride staining and by the expression of several adipocyte markers: aP2, adipsin, C/EBPalpha, and Pref-1. Moreover, cyclopamine cannot substitute for other components of the differentiation medium: insulin, dexamethasone or IBMX. These results indicate that although Hedgehog signaling decreases during adipocyte differentiation, this down-regulation is not sufficient to trigger adipocyte differentiation. This suggests that Hedgehog signaling is an inadequate pharmacological target for patient suffering from syndromes associated with a decrease in fat mass, such as the ones observed in lipodystrophies.
Collapse
Affiliation(s)
- W Cousin
- Institute of Signaling, Developmental Biology and Cancer CNRS UMR6543, Université de Nice Sophia-Antipolis, Stem Cells and Differentiation, Nice, France
| | | | | |
Collapse
|
41
|
De Rivoyre M, Ruel L, Varjosalo M, Loubat A, Bidet M, Thérond P, Mus-Veteau I. Human receptors patched and smoothened partially transduce hedgehog signal when expressed in Drosophila cells. J Biol Chem 2006; 281:28584-95. [PMID: 16867986 DOI: 10.1074/jbc.m512986200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In humans, dysfunctions of the Hedgehog receptors Patched and Smoothened are responsible for numerous pathologies. However, signaling mechanisms involving these receptors are less well characterized in mammals than in Drosophila. To obtain structure-function relationship information on human Patched and Smoothened, we expressed these human receptors in Drosophila Schneider 2 cells. We show here that, as its Drosophila counterpart, human Patched is able to repress the signaling pathway in the absence of Hedgehog ligand. In response to Hedgehog, human Patched is able to release Drosophila Smoothened inhibition, suggesting that human Patched is expressed in a functional state in Drosophila cells. We also provide experiments showing that human Smo, when expressed in Schneider cells, is able to bind the alkaloid cyclopamine, suggesting that it is expressed in a native conformational state. Furthermore, contrary to Drosophila Smoothened, human Smoothened does not interact with the kinesin Costal 2 and thus is unable to transduce the Hedgehog signal. Moreover, cell surface fluorescent labeling suggest that human Smoothened is enriched at the Schneider 2 plasma membrane in response to Hedgehog. These results suggest that human Smoothened is expressed in a functional state in Drosophila cells, where it undergoes a regulation of its localization comparable with its Drosophila homologue. Thus, we propose that the upstream part of the Hedgehog pathway involving Hedgehog interaction with Patched, regulation of Smoothened by Patched, and Smoothened enrichment at the plasma membrane is highly conserved between Drosophila and humans; in contrast, signaling downstream of Smoothened is different.
Collapse
Affiliation(s)
- Matthieu De Rivoyre
- Laboratoire de Physiologie Cellulaire et Moléculaire, CNRS Unité Mixte de Recherche (UMR) 6548, Université de Nice-Sophia Antipolis, Parc Valrose 06108 Nice Cedex 2, France
| | | | | | | | | | | | | |
Collapse
|
42
|
Loulier K, Ruat M, Traiffort E. Increase of proliferating oligodendroglial progenitors in the adult mouse brain upon Sonic hedgehog delivery in the lateral ventricle. J Neurochem 2006; 98:530-42. [PMID: 16805844 DOI: 10.1111/j.1471-4159.2006.03896.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Sonic hedgehog signaling is required for the maintenance of stem cell niches in the postnatal subventricular zone and the proliferation of neural progenitors in the mature hippocampus. We show here that delivery of Sonic hedgehog protein into the lateral ventricle of adult mice increases cell proliferation in the corpus callosum and cerebral cortex. In this latter area, the number of neural progenitors expressing the proteoglycan NG2 is enhanced 2 days after the injection. In both areas, mRNA up-regulation of the transcriptional target gene Patched was observed in cells expressing the oligodendroglial transcription factor Olig1. Twenty-six days following the adenovirus-mediated delivery of Sonic hedgehog into the lateral ventricle, newly generated cells in the cerebral cortex and in the corpus callosum are influenced towards the initial steps of oligodendrogenesis, as indicated by a 50% increase in the number of cells expressing the oligodendroglial marker DM20. Our experiments demonstrate that the number of oligodendrocyte precursor cells in the cerebral cortex and corpus callosum can be increased upon delivery of Sonic hedgehog proteins and highlight the potential capacity of the adult brain to mobilize a pool of premyelinating cells.
Collapse
Affiliation(s)
- Karine Loulier
- CNRS, Signal Transduction and Developmental Neuropharmacology, UPR9040 Laboratoire de Neurobiologie Cellulaire et Moléculaire, Institut de Neurobiologie Alfred Fessard, IFR 2118, Gif sur Yvette, France
| | | | | |
Collapse
|
43
|
Lipinski RJ, Gipp JJ, Zhang J, Doles JD, Bushman W. Unique and complimentary activities of the Gli transcription factors in Hedgehog signaling. Exp Cell Res 2006; 312:1925-38. [PMID: 16571352 DOI: 10.1016/j.yexcr.2006.02.019] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Revised: 02/15/2006] [Accepted: 02/22/2006] [Indexed: 10/24/2022]
Abstract
The Gli family of transcription factors (Gli1, 2 and 3) mediate the Hedgehog morphogenetic signal by regulating the expression of downstream target genes. Aberrations in Hedgehog signaling seriously affect vertebrate development. Postnatally, Hedgehog signaling has been postulated to play a pivotal role in healing and repair processes and inappropriate pathway activation has been implicated in several types of cancers. To better understand both the upstream regulation of the Gli transcription factors, as well as their unique and combinatorial roles in regulating the expression of Hedgehog target genes, we have characterized embryonic fibroblasts (MEFs) from Gli mutant mice. Stimulation of wild-type MEFs by Sonic Hedgehog (Shh) peptide elicited unique profiles of induction of Hedgehog target genes Gli1, Ptc1, and Hip1. Gli2 loss-of-function was associated with diminished Shh-induced target gene expression, while Gli3 loss-of-function was associated with increased basal and Shh-induced target gene expression. The loss of Gli1 alone had no effect on target gene induction but did diminish Shh-induced target gene expression when combined with the loss of Gli2 or Gli3. Additionally, overexpression of Gli1 induced target gene expression in Gli2(-/-)3(-/-) MEFs, while Shh stimulation did not. Using MEFs expressing only Gli2 or Gli3, we found that both cyclopamine and the PKA activator forskolin inhibited target gene induction mediated by Gli2 and Gli3. These results demonstrate that Gli2 and Gli3 share common regulatory mechanisms and modulate Hedgehog target gene expression directly and independently while also regulating Gli1 expression, which in specific contexts, coordinately contributes to target gene activation.
Collapse
Affiliation(s)
- Robert J Lipinski
- Molecular and Environmental Toxicology Center, Madison, WI 53705-222, USA
| | | | | | | | | |
Collapse
|
44
|
Østerlund T, Kogerman P. Hedgehog signalling: how to get from Smo to Ci and Gli. Trends Cell Biol 2006; 16:176-80. [PMID: 16516476 DOI: 10.1016/j.tcb.2006.02.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Revised: 02/02/2006] [Accepted: 02/17/2006] [Indexed: 11/21/2022]
Abstract
The secreted morphogens of the Hedgehog family have important roles in normal development as well as in associated pathologies, including cancer. The Hedgehog signalling pathway has been studied in Drosophila and is thought to be conserved in vertebrates. Hedgehog elicits a signalling response that activates Smoothened (Smo). There is evidence of differences between Drosophila and vertebrates concerning signalling downstream of Smo, as well as in Smo itself. Here, we discuss this evidence and its importance for investigations of the pathway and related biology, as well as for the development of drugs targeting components of the pathway for treatment of associated pathologies.
Collapse
Affiliation(s)
- Torben Østerlund
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia.
| | | |
Collapse
|
45
|
Chen MH, Gao N, Kawakami T, Chuang PT. Mice deficient in the fused homolog do not exhibit phenotypes indicative of perturbed hedgehog signaling during embryonic development. Mol Cell Biol 2005; 25:7042-53. [PMID: 16055716 PMCID: PMC1190231 DOI: 10.1128/mcb.25.16.7042-7053.2005] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hedgehog (Hh) signaling plays a major role in multiple aspects of embryonic development. To understand how a single Hh signal is capable of generating distinct readouts in Hh-responsive cells requires elucidation of the signal transduction cascade at the molecular level. Key components that mediate Hh signal transduction downstream of the receptor include Fused (Fu), Suppressor of fused (Sufu), and Costal-2 (Cos2) or the vertebrate homologs Kif27/Kif7. Studies with both invertebrates and vertebrates have led to a model in which a protein complex composed of Fu, Sufu, and Cos2 controls the processing, activity, and subcellular distribution of the Ci/Gli transcription factors responsible for Hh target gene activation. These converging results obtained with different species reaffirm the prevailing view of pathway conservation during evolution. Genetic studies of Fu, Sufu, and Kif27/Kif7 in mice are required to provide further verification of Hh pathway conservation. To this end, we generated a gene-targeted allele of Fu in mice. Surprisingly, our analysis indicates that Fu-deficient mice do not exhibit any embryonic phenotypes indicative of perturbed Hh signaling. This could be due to either functional redundancy or Hh pathway divergence and clearly indicates greater complexity of Hh signaling in vertebrates.
Collapse
Affiliation(s)
- Miao-Hsueh Chen
- Cardiovascular Research Institute, University of California, San Francisco, 94143, USA
| | | | | | | |
Collapse
|