201
|
Wang Y, Imitola J, Rasmussen S, O’Connor KC, Khoury SJ. Paradoxical dysregulation of the neural stem cell pathway sonic hedgehog-Gli1 in autoimmune encephalomyelitis and multiple sclerosis. Ann Neurol 2008; 64:417-27. [PMID: 18991353 PMCID: PMC2757750 DOI: 10.1002/ana.21457] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Neurovascular niches have been proposed as critical components of the neural stem cell (NSC) response to acute central nervous system injury; however, it is unclear whether these potential reparative niches remain functional during chronic injury. Here, we asked how central nervous system inflammatory injury regulates the intrinsic properties of NSCs and their niches. METHODS We investigated the sonic hedgehog (Shh)-Gli1 pathway, an important signaling pathway for NSCs, in experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS), and its regulation by inflammatory cytokines. RESULTS We show that Shh is markedly upregulated by reactive and perivascular astroglia in areas of injury in MS lesions and during EAE. Astroglia outside the subventricular zone niche can support NSC differentiation toward neurons and oligodendrocytes, and Shh is a critical mediator of this effect. Shh induces differential upregulation of the transcription factor Gli1, which mediates Shh-induced NSC differentiation. However, despite the increase in Shh and the fact that Gli1 was initially increased during early inflammation of EAE and active lesions of MS, Gli1 was significantly decreased in spinal cord oligodendrocyte precursor cells after onset of EAE, and in chronic active and inactive lesions from MS brain. The Th1 cytokine interferon-gamma was unique in inducing Shh expression in astroglia and NSCs, while paradoxically suppressing Gli1 expression in NSCs and inhibiting Shh-mediated NSC differentiation. INTERPRETATION Our data suggest that endogenous repair potential during chronic injury appears to be limited by inflammation-induced alterations in intrinsic NSC molecular pathways such as Gli1.
Collapse
MESH Headings
- Animals
- Astrocytes/physiology
- Cell Differentiation/physiology
- Cells, Cultured
- Cerebral Cortex/cytology
- Cerebral Cortex/pathology
- Embryo, Mammalian
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Green Fluorescent Proteins/biosynthesis
- Green Fluorescent Proteins/genetics
- Hedgehog Proteins/metabolism
- Humans
- Interferon-gamma/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/pathology
- Neurons/physiology
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Receptors, Antigen, T-Cell/genetics
- Stem Cells/drug effects
- Stem Cells/physiology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Up-Regulation/drug effects
- Up-Regulation/physiology
- Zinc Finger Protein GLI1
Collapse
Affiliation(s)
- Yue Wang
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jaime Imitola
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Stine Rasmussen
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Anatomy and Neurobiology, University of Southern Denmark, DK-5000 Odense C, Denmark
| | - Kevin C. O’Connor
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Samia J. Khoury
- Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
202
|
Xu Q, Yuan X, Liu G, Black KL, Yu JS. Hedgehog signaling regulates brain tumor-initiating cell proliferation and portends shorter survival for patients with PTEN-coexpressing glioblastomas. Stem Cells 2008; 26:3018-26. [PMID: 18787206 DOI: 10.1634/stemcells.2008-0459] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The identification of brain tumor stem-like cells (BTSCs) has implicated a role of biological self-renewal mechanisms in clinical brain tumor initiation and propagation. The molecular mechanisms underlying the tumor-forming capacity of BTSCs, however, remain unknown. Here, we have generated molecular signatures of glioblastoma multiforme (GBM) using gene expression profiles of BTSCs and have identified both Sonic Hedgehog (SHH) signaling-dependent and -independent BTSCs and their respective glioblastoma surgical specimens. BTSC proliferation could be abrogated in a pathway-dependent fashion in vitro and in an intracranial tumor model in athymic mice. Both SHH-dependent and -independent brain tumor growth required phosphoinositide 3-kinase-mammalian target of rapamycin signaling. In human GBMs, the levels of SHH and PTCH1 expression were significantly higher in PTEN-expressing tumors than in PTEN-deficient tumors. In addition, we show that hyperactive SHH-GLI signaling in PTEN-coexpressing human GBM is associated with reduced survival time. Thus, distinct proliferation signaling dependence may underpin glioblastoma propagation by BTSCs. Modeling these BTSC proliferation mechanisms may provide a rationale for individualized glioblastoma treatment.
Collapse
Affiliation(s)
- Qijin Xu
- Maxine Dunitz Neurosurgical Institute, Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California 90049, USA
| | | | | | | | | |
Collapse
|
203
|
Mash1 and neurogenin 2 enhance survival and differentiation of neural precursor cells after transplantation to rat brains via distinct modes of action. Mol Ther 2008; 16:1873-82. [PMID: 18781144 DOI: 10.1038/mt.2008.189] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Neural precursor cells (NPCs) are regarded as a promising source of donor cells in transplantation-based therapies for neurodegenerative disorders. However, poor survival and limited neuronal differentiation of the transplanted NPCs remain critical limitations for developing therapeutic strategies. In this study, we investigated the effects of the proneural basic helix-loop-helix (bHLH) transcription factors Mash1 and Neurogenin 2 (Ngn2) in neuronal differentiation and survival of NPCs. Induction of Mash1 or Ngn2 expression strikingly enhanced neuronal differentiation of cultured NPCs in vitro. Ngn2-transduced NPCs underwent a rapid cell cycle arrest, which often accompanies differentiation. In contrast, cells continuously expanded upon Mash1 expression during NPC differentiation. Notably, sonic hedgehog (SHH) was upregulated by Mash1 and mediated the proliferative and survival effects of Mash1 on NPCs. Upon transplantation into adult rat brains, Mash1-expressing NPCs yielded large grafts enriched with neurons compared to control LacZ-transduced NPCs. Interestingly, enhancements in neuronal yield, as well as in donor cell survival, were also achieved by transplanting Ngn2-transduced NPCs. We show that a differentiation stage- and cell density-dependent survival effect of Ngn2 involves neurotrophin3 (NT3)/TrkC-mediated signaling. Together, these findings suggest potential benefits of bHLH gene manipulation to develop successful transplantation strategies for brain disorders.
Collapse
|
204
|
Abstract
Brain tumors are generally incurable cancers. Work from a number of laboratories strongly suggests that they are organized as a hierarchy based on a subset of cancer cells that have stem-cell properties. These cells have now been shown to be resistant to conventional therapy and responsive to differentiation therapy. New in vitro and in vivo models for interrogating brain tumor cells in stem-cell conditions have been developed that provide important new opportunities for elucidating the key pathways responsible for driving the proliferation of these cells. Continued application of the principles of stem-cell biology to the study of brain cancers is likely to continue to bring further important insight into these aggressive cancers, bringing new treatments and understanding of the origins.
Collapse
Affiliation(s)
- Peter B Dirks
- Arthur and Sonia Labatt Brain Tumor Research Center, Program in Developmental and Stem Cell Biology, Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, Ontario, Canada.
| |
Collapse
|
205
|
Abstract
Medulloblastoma and other embronal brain tumors are similar in appearance and differentiation potential to neural stem and progenitor cells. Expression studies performed using human tumor samples, as well as the analysis of murine transgenic models, suggest that both multipotent cerebellar stem cells and lineage-restricted progenitors of the external germinal layer can be transformed into medulloblastoma by genetic alterations. These molecular changes frequently involve constitutive activation of signaling pathways such as Wnt, Hedgehog, and Notch, which play a key role in non-neoplastic neural stem cells. Pharmacologic blockade of the Hedgehog and Notch pathways suppresses the growth of medulloblastoma in culture and in vivo and may prove effective in targeting the small cancer stem-cell subpopulation required for tumor initiation and long-term propagation.
Collapse
Affiliation(s)
- Xing Fan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
206
|
Zhao H, Ayrault O, Zindy F, Kim JH, Roussel MF. Post-transcriptional down-regulation of Atoh1/Math1 by bone morphogenic proteins suppresses medulloblastoma development. Genes Dev 2008; 22:722-7. [PMID: 18347090 DOI: 10.1101/gad.1636408] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Bone morphogenic proteins 2 and 4 (BMP2 and BMP4) inhibit proliferation and induce differentiation of cerebellar granule neuron progenitors (GNPs) and primary GNP-like medulloblastoma (MB) cells. This occurs through rapid proteasome-mediated degradation of Math1 (Atoh1), a transcription factor expressed in proliferating GNPs. Ectopic expression of Atoh1, but not of Sonic hedgehog (Shh)-regulated Gli1 or Mycn, cancels these BMP-mediated effects and restores Shh-dependent proliferation of GNPs and MB cells in vitro and in vivo. Genes regulating the BMP signaling pathway are down-regulated in mouse MBs. Thus, BMPs are potent inhibitors of MB and should be considered as novel therapeutic agents.
Collapse
Affiliation(s)
- Haotian Zhao
- Departments of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | |
Collapse
|
207
|
Hedgehog Serves as a Mitogen and Survival Factor During Embryonic Stem Cell Neurogenesis. Stem Cells 2008; 26:1097-108. [DOI: 10.1634/stemcells.2007-0684] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
208
|
Becher OJ, Hambardzumyan D, Fomchenko EI, Momota H, Mainwaring L, Bleau AM, Katz AM, Edgar M, Kenney AM, Cordon-Cardo C, Blasberg RG, Holland EC. Gli activity correlates with tumor grade in platelet-derived growth factor-induced gliomas. Cancer Res 2008; 68:2241-9. [PMID: 18381430 DOI: 10.1158/0008-5472.can-07-6350] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gli signaling is critical for central nervous system development and is implicated in tumorigenesis. To monitor Gli signaling in gliomas in vivo, we created platelet-derived growth factor-induced gliomas in a Gli-luciferase reporter mouse. We find that Gli activation is found in gliomas and correlates with grade. In addition, we find that sonic hedgehog (SHH) is expressed in these tumors and also correlates with grade. We identify microvascular proliferation and pseudopalisades, elements that define high-grade gliomas as SHH-producing microenvironments. We describe two populations of SHH-producing stromal cells that reside in perivascular niche (PVN), namely low-cycling astrocytes and endothelial cells. Using the Ptc-LacZ knock-in mouse as a second Gli responsive reporter, we show beta-galactosidase activity in the PVN and in some tumors diffusely throughout the tumor. Lastly, we observe that SHH is similarly expressed in human gliomas and note that an intact tumor microenvironment or neurosphere conditions in vitro are required for Gli activity.
Collapse
Affiliation(s)
- Oren J Becher
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Dirks PB. Brain tumour stem cells: the undercurrents of human brain cancer and their relationship to neural stem cells. Philos Trans R Soc Lond B Biol Sci 2008; 363:139-52. [PMID: 17309866 PMCID: PMC2605491 DOI: 10.1098/rstb.2006.2017] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Conceptual and technical advances in neural stem cell biology are being applied to the study of human brain tumours. These studies suggest that human brain tumours are organized as a hierarchy and are maintained by a small number of tumour cells that have stem cell properties. Most of the bulk population of human brain tumours comprise cells that have lost the ability to initiate and maintain tumour growth. Although the cell of origin for human brain tumours is uncertain, recent evidence points towards the brain's known proliferative zones. The identification of brain tumour stem cells has important implications for understanding brain tumour biology and these cells may be critical cellular targets for curative therapy.
Collapse
Affiliation(s)
- Peter B Dirks
- Division of Neurosurgery and Program in Developmental Biology, University of Toronto, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada.
| |
Collapse
|
210
|
Glatt S, Halbauer D, Heindl S, Wernitznig A, Kozina D, Su KC, Puri C, Garin-Chesa P, Sommergruber W. hGPR87 contributes to viability of human tumor cells. Int J Cancer 2008; 122:2008-16. [PMID: 18183596 DOI: 10.1002/ijc.23349] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Emerging in vitro and in vivo data underline the crucial role of G-protein-coupled receptors (GPCRs) in tumorigenesis. Here, we report the contribution of hGPR87, a predicted member of the P2Y subfamily of GPCRs, to proliferation and survival of human tumor cell lines. hGPR87 mRNA transcript was found to be preferentially overexpressed in squamous cell carcinomas (SCCs) of different locations and in their lymph node metastases. Up-regulation of both, transcript and protein, was detected in samples of SCC of the lung, cervix, skin and head and neck (pharynx, larynx and epiglottis). In addition to the expression of hGPR87 in tumors which originate from stratified epithelia, we identified other hGPR87-positive tumor types including subsets of large cell and adenocarcinomas of the lung and transitional cell carcinomas of the urinary bladder. Loss of function studies using siRNA in human cancer cell lines lead to antiproliferative effects and induction of apoptosis. Like other known P2Y receptors, hGPR87 was found to be mainly located on the cell surface. The overexpression of hGPR87 preferentially in SCCs together with our functional data suggests a common molecular mechanism for SCC tumorigenesis and may provide a novel intervention site for mechanism-based antitumor therapies.
Collapse
Affiliation(s)
- Sebastian Glatt
- Boehringer-Ingelheim Austria GmbH, Dr. Boehringer-Gassse 5-11, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
211
|
The Smo/Smo Model: Hedgehog-Induced Medulloblastoma with 90% Incidence and Leptomeningeal Spread. Cancer Res 2008; 68:1768-76. [DOI: 10.1158/0008-5472.can-07-5092] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
212
|
Galvin KE, Ye H, Erstad DJ, Feddersen R, Wetmore C. Gli1 induces G2/M arrest and apoptosis in hippocampal but not tumor-derived neural stem cells. Stem Cells 2008; 26:1027-36. [PMID: 18276799 DOI: 10.1634/stemcells.2007-0879] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Sonic hedgehog (Shh) is necessary for sustaining the proliferation of neural stem cells (NSCs), yet little is known about its mechanisms. Whereas Gli1, Gli2, and Gli3, the primary mediators of Shh signaling, were all expressed in hippocampal neural progenitors, Shh treatment of NSCs induced only Gli1 expression. Acute depletion of Gli1 in postnatal NSCs by short-hairpin RNA decreased proliferation, whereas germline deletion of Gli1 did not affect NSC proliferation, suggesting a difference in mechanisms of Gli1 compensation that may be developmentally dependent. To determine whether Gli1 was sufficient to enhance NSC proliferation, we overexpressed this mitogen and were surprised to find that Gli1 resulted in decreased proliferation, accumulation of NSCs in the G2/M phase of cell cycle, and apoptosis. In contrast, Gli1-expressing lineage-restricted neural precursors demonstrated a 4.5-fold proliferation enhancement. Expression analyses of Gli1-expressing NSCs identified significant induction of Gadd45a and decreased cyclin A2 and Stag1 mRNA, genes involved in the G2-M transition and apoptosis. Furthermore, Gadd45a overexpression was sufficient to partially recapitulate the Gli1-induced G2/M accumulation and cell death of NSCs. In contrast to normal stem cells, tumor-derived stem cells had markedly higher basal Gli1 expression and did not undergo apoptosis with further elevation of Gli1. Our data suggest that Gli1-induced apoptosis may serve as a protective mechanism against premature mitosis and may give insight into mechanisms by which nonmalignant stem cells restrain hyperproliferation in the context of potentially transforming mitogenic signals. Tumor-derived stem cells apparently lack these mechanisms, which may contribute to their unrestrained proliferation and malignant potential.
Collapse
Affiliation(s)
- Katherine E Galvin
- Program in Molecular Neuroscience, The College of Idaho, Caldwell, Idaho, USA
| | | | | | | | | |
Collapse
|
213
|
Sauvageot CM, Kesari S, Stiles CD. Molecular pathogenesis of adult brain tumors and the role of stem cells. Neurol Clin 2008; 25:891-924, vii. [PMID: 17964020 DOI: 10.1016/j.ncl.2007.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Primary brain tumors consist of neoplasms with varied molecular defects, morphologic phenotypes, and clinical outcomes. The genetic and signaling abnormalities involved in tumor initiation and progression of the most prevalent adult primary brain tumors, including gliomas, meningiomas, and medulloblastomas, are described in this article. The current understanding of the cell-of-origin of these neoplasms is reviewed, which suggests that the malignant phenotype is propelled by cells with stem-like qualities. A comprehensive understanding of the molecular basis of transformation and the cell-of-origin of these neoplasms will enable the formulation of more targeted treatment alternatives that could improve survival and quality of life.
Collapse
Affiliation(s)
- Claire M Sauvageot
- Department of Cancer Biology, Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115-6084, USA
| | | | | |
Collapse
|
214
|
Fernandez C, Soulier M, Coulibaly B, Liprandi A, Benoit B, Giuliano F, Sigaudy S, Figarella-Branger D, Fallet-Bianco C. Acrocallosal syndrome in fetus: focus on additional brain abnormalities. Acta Neuropathol 2008; 115:151-6. [PMID: 17593378 DOI: 10.1007/s00401-007-0249-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2007] [Revised: 05/24/2007] [Accepted: 06/04/2007] [Indexed: 10/23/2022]
Abstract
Acrocallosal syndrome (ACS) is an autosomal recessive disorder characterized by craniofacial dysmorphism, agenesis or hypoplasia of the corpus callosum, duplication of the phalanges of the hallux, more rarely the thumbs, post-axial polydactyly, syndactyly and severe mental retardation. Here we report the two first descriptions of acrocallosal syndrome in fetus with extensive neuropathological study and provide new data regarding additional brain abnormalities in ACS. The first case was a 25-gestational week male fetus displaying craniofacial and limb abnormalities, with bilateral syndactyly of the fourth and fifth fingers, preaxial polydactyly of the left foot and an inter-frontal extra-bone. The second fetus was a 33-gestational week male fetus. His left hand displayed a broad thumb and 4/5 syndactyly. In both cases, gross examination of the brain showed an absence of corpus callosum associated with interhemispheric cysts. The cerebral cortex in front of the cysts was nodular. Upon microscopic examination, the nodular masses corresponded to large dysplastic areas represented by clusters of undifferentiated neurons in the white matter. The cyst wall showed arachnoidal and ependymal covering and contained numerous choroid plexus, suggesting a developmental abnormality of the ventricles. The pons and the cerebellum were hypoplastic. The dentate nuclei were fragmented. Numerous neuronal heterotopias associated with ectopic ependymal cavities were observed in the vermis in one case. The olivary nuclei were severely dysplastic too. We hope that these new data will make both the ante- and post-natal diagnosis easier, facilitate comparisons with animal models and encourage the identification of the genes responsible for this syndrome.
Collapse
Affiliation(s)
- Carla Fernandez
- Laboratoire d'Anatomie Pathologique et Neuropathologie, Hôpital de la Timone Adultes, 264 rue Saint-Pierre, 13385, Marseille Cedex 05, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Hayhurst M, Gore BB, Tessier-Lavigne M, McConnell SK. Ongoing sonic hedgehog signaling is required for dorsal midline formation in the developing forebrain. Dev Neurobiol 2008; 68:83-100. [DOI: 10.1002/dneu.20576] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
216
|
Diabira S, Morandi X. Gliomagenesis and neural stem cells: Key role of hypoxia and concept of tumor “neo-niche”. Med Hypotheses 2008; 70:96-104. [PMID: 17614215 DOI: 10.1016/j.mehy.2007.04.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Accepted: 04/16/2007] [Indexed: 11/17/2022]
Abstract
Gliomas represent the most common primary brain tumors and the most devastating pathology of the central nervous system. Despite progress in conventional treatments, the prognosis remains dismal. Recent studies have suggested that a glioma brain tumor may arise from a "cancer stem cell". To understand this theory we summarize studies of the concepts of neural stem cell, and its specialized microenvironment, namely the niche which can regulate balanced self-renewal, differentiation and stem cell quiescence. We summarize the molecular mechanism known or postulated to be involved in the disregulation of normal stem cells features allowing them to undergo neoplasic transformation. We seek data pointing out the key role of hypoxia in normal homeostasis of stem cells and in the initiation, development and aggressiveness of gliomas. We develop the concept of tumor special microenvironment and we propose the new concept of neo-niche, surrounding the glioma, in which hypoxia could be a key factor to recruit and deregulate different stem cells for gliogenesis process. Substantial advances in treatment would come from obtaining better knowledge of molecular impairs of this disease.
Collapse
Affiliation(s)
- Sylma Diabira
- Department of Neurosurgery, CHRU Pontchaillou, Rue Henri Le Guilloux, 35033 Rennes, Cedex 2, France.
| | | |
Collapse
|
217
|
Barami K. Biology of the subventricular zone in relation to gliomagenesis. J Clin Neurosci 2007; 14:1143-9. [DOI: 10.1016/j.jocn.2007.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 04/02/2007] [Accepted: 04/03/2007] [Indexed: 01/05/2023]
|
218
|
Hatton BA, Knoepfler PS, Kenney AM, Rowitch DH, de Alborán IM, Olson JM, Eisenman RN. N-myc is an essential downstream effector of Shh signaling during both normal and neoplastic cerebellar growth. Cancer Res 2007; 66:8655-61. [PMID: 16951180 DOI: 10.1158/0008-5472.can-06-1621] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We examined the genetic requirements for the Myc family of oncogenes in normal Sonic hedgehog (Shh)-mediated cerebellar granule neuronal precursor (GNP) expansion and in Shh pathway-induced medulloblastoma formation. In GNP-enriched cultures derived from N-myc(Fl/Fl) and c-myc(Fl/Fl) mice, disruption of N-myc, but not c-myc, inhibited the proliferative response to Shh. Conditional deletion of c-myc revealed that, although it is necessary for the general regulation of brain growth, it is less important for cerebellar development and GNP expansion than N-myc. In vivo analysis of compound mutants carrying the conditional N-myc null and the activated Smoothened (ND2:SmoA1) alleles showed, that although granule cells expressing the ND2:SmoA1 transgene are present in the N-myc null cerebellum, no hyperproliferation or tumor formation was detected. Taken together, these findings provide in vivo evidence that N-myc acts downstream of Shh/Smo signaling during GNP proliferation and that N-myc is required for medulloblastoma genesis even in the presence of constitutively active signaling from the Shh pathway.
Collapse
Affiliation(s)
- Beryl A Hatton
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA 98109, USA.
| | | | | | | | | | | | | |
Collapse
|
219
|
Antón Aparicio LM, García Campelo R, Cassinello Espinosa J, Valladares Ayerbes M, Reboredo López M, Díaz Prado S, Aparicio Gallego G. Prostate cancer and Hedgehog signalling pathway. Clin Transl Oncol 2007; 9:420-8. [PMID: 17652055 DOI: 10.1007/s12094-007-0080-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The Hedgehog (Hh) family of intercellular signalling proteins have come to be recognised as key mediators in many fundamental processes in embryonic development. Their activities are central to the growth, patterning and morphogenesis of many different regions within the bodies of vertebrates. In some contexts, Hh signals act as morphogens in the dose-dependent induction of distinct cell fates within a target field, in others as mitogens in the regulation of cell proliferation or as inducing factors controlling the form of a developing organ. These diverse functions of Hh proteins raise many intriguing questions about their mode of action. Various studies have now demonstrated the function of Hh signalling in the control of cell proliferation, especially for stem cells and stem-like progenitors. Abnormal activation of the Hh pathway has been demonstrated in a variety of human tumours. Hh pathway activity in these tumours is required for cancer cell proliferation and tumour growth. Recent studies have uncovered the role for Hh signalling in advanced prostate cancer and demonstrated that autocrine signalling by tumour cells is required for proliferation, viability and invasive behaviour. Thus, Hh signalling represents a novel pathway in prostate cancer that offers opportunities for prognostic biomarker development, drug targeting and therapeutic response monitoring.
Collapse
Affiliation(s)
- L M Antón Aparicio
- Medical Oncology Service, C.H.U. Juan Canalejo, Department of Medicine, University of La Coruña, A Coruña, Spain.
| | | | | | | | | | | | | |
Collapse
|
220
|
Wan J, Zheng H, Xiao HL, She ZJ, Zhou GM. Sonic hedgehog promotes stem-cell potential of Müller glia in the mammalian retina. Biochem Biophys Res Commun 2007; 363:347-54. [PMID: 17880919 DOI: 10.1016/j.bbrc.2007.08.178] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 08/27/2007] [Indexed: 10/22/2022]
Abstract
Müller glia have been demonstrated to display stem-cell properties after retinal damage. Here, we report this potential can be regulated by Sonic hedgehog (Shh) signaling. Shh can stimulate proliferation of Müller glia through its receptor and target gene expressed on them, furthermore, Shh-treated Müller glia are induced to dedifferentiate by expressing progenitor-specific markers, and then adopt cell fate of rod photoreceptor. Inhibition of signaling by cyclopamine inhibits proliferation and dedifferentiation. Intraocular injection of Shh promotes Müller glia activation in the photoreceptor-damaged retina, Shh also enhances neurogenic potential by producing more rhodopsin-positive photoreceptors from Müller glia-derived cells. Together, these results provide evidences that Müller glia act as potential stem cells in mammalian retina, Shh may have therapeutic effects on these cells for promoting the regeneration of retinal neurons.
Collapse
Affiliation(s)
- Jin Wan
- Department of Anatomy, Histology and Embryology, Shanghai Medical School, Fudan University, 200032 Shanghai, China
| | | | | | | | | |
Collapse
|
221
|
Bar EE, Chaudhry A, Lin A, Fan X, Schreck K, Matsui W, Piccirillo S, Vescovi AL, Dimeco F, Olivi A, Eberhart CG. Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like cancer cells in glioblastoma. Stem Cells 2007; 25:2524-33. [PMID: 17628016 PMCID: PMC2610257 DOI: 10.1634/stemcells.2007-0166] [Citation(s) in RCA: 450] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Brain tumors can arise following deregulation of signaling pathways normally activated during brain development and may derive from neural stem cells. Given the requirement for Hedgehog in non-neoplastic stem cells, we investigated whether Hedgehog blockade could target the stem-like population in glioblastoma multiforme (GBM). We found that Gli1, a key Hedgehog pathway target, was highly expressed in 5 of 19 primary GBM and in 4 of 7 GBM cell lines. Shh ligand was expressed in some primary tumors, and in GBM-derived neurospheres, suggesting a potential mechanism for pathway activation. Hedgehog pathway blockade by cyclopamine caused a 40%-60% reduction in growth of adherent glioma lines highly expressing Gli1 but not in those lacking evidence of pathway activity. When GBM-derived neurospheres were treated with cyclopamine and then dissociated and seeded in media lacking the inhibitor, no new neurospheres formed, suggesting that the clonogenic cancer stem cells had been depleted. Consistent with this hypothesis, the stem-like fraction in gliomas marked by both aldehyde dehydrogenase activity and Hoechst dye excretion (side population) was significantly reduced or eliminated by cyclopamine. In contrast, we found that radiation treatment of our GBM neurospheres increased the percentage of these stem-like cells, suggesting that this standard therapy preferentially targets better-differentiated neoplastic cells. Most importantly, viable GBM cells injected intracranially following Hedgehog blockade were no longer able to form tumors in athymic mice, indicating that a cancer stem cell population critical for ongoing growth had been removed. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Eli E. Bar
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aneeka Chaudhry
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alex Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xing Fan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karisa Schreck
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William Matsui
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sara Piccirillo
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | - Angelo L. Vescovi
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | | | - Alessandro Olivi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles G. Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
222
|
Ruiz i Altaba A, Mas C, Stecca B. The Gli code: an information nexus regulating cell fate, stemness and cancer. Trends Cell Biol 2007; 17:438-47. [PMID: 17845852 PMCID: PMC2601665 DOI: 10.1016/j.tcb.2007.06.007] [Citation(s) in RCA: 302] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 06/30/2007] [Accepted: 06/30/2007] [Indexed: 12/11/2022]
Abstract
The Gli code hypothesis postulates that the three vertebrate Gli transcription factors act together in responding cells to integrate intercellular Hedgehog (Hh) and other signaling inputs, resulting in the regulation of tissue pattern, size and shape. Hh and other inputs are then just ways to modify the Gli code. Recent data confirm this idea and suggest that the Gli code regulates stemness and also tumor progression and metastatic growth, opening exciting possibilities for both regenerative medicine and novel anticancer therapies.
Collapse
Affiliation(s)
- Ariel Ruiz i Altaba
- Department of Genetic Medicine and Development, 8242 CMU, University of Geneva Medical School, 1 rue Michel Servet, CH-1211 Geneva, Switzerland.
| | | | | |
Collapse
|
223
|
Hu WG, Liu T, Xiong JX, Wang CY. Blockade of sonic hedgehog signal pathway enhances antiproliferative effect of EGFR inhibitor in pancreatic cancer cells. Acta Pharmacol Sin 2007; 28:1224-30. [PMID: 17640486 DOI: 10.1111/j.1745-7254.2007.00620.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM To investigate the expression of sonic hedgehog (SHH) and epidermal growth factor receptor (EGFR) signal molecules in pancreatic cancer cells, and to assess the inhibitory effects through the blockade of the SHH and EGFR signaling pathways by cyclopamine and Iressa, respectively. METHODS The expression of SHH and EGFR in pancreatic cancer cell lines (PANC-1, SUIT-2, and ASPC-1) was detected by RT-PCR and Western blot analysis. After treatment with different concentrations of cyclopamine, alone or in combination with Iressa, the antiproliferative effect on pancreatic cancer cells was analyzed by methyl thiazolyl tetrazolium assays. A flow cytometry analysis was used to detect the cellular cycle distribution and apoptosis of pancreatic cancer cells. RESULTS All of the 3 pancreatic cancer cell lines expressed SHH, Smoothened (SMO), and EGFR. Cyclopamine could downregulate the expression of EGFR in all cell lines. Cyclopamine or Iressa could induce a growth inhibitory effect in a dose-dependent manner. Moreover, the combined use of 2.5 micromol/L cyclopamine and 1 micromol/L Iressa induced an enhanced inhibitory effect and a greater apoptosis rate than any agent alone. The percentage of the cell population of the G0/G1 and sub-G1 phases was significantly increased along with the increasing dose of cyclopamine and/or Iressa. CONCLUSION The blockade of the sonic hedgehog signal pathway enhances the antiproliferative effect of the EGFR inhibitor through the downregulation of its expression in pancreatic cancer cells. The simultaneous blockade of SHH and EGFR signaling represents possible targets of new treatment strategies for pancreatic carcinoma.
Collapse
Affiliation(s)
- Wei-guo Hu
- Pancreatic Surgery Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | |
Collapse
|
224
|
Ma YH, Mentlein R, Knerlich F, Kruse ML, Mehdorn HM, Held-Feindt J. Expression of stem cell markers in human astrocytomas of different WHO grades. J Neurooncol 2007; 86:31-45. [PMID: 17611714 DOI: 10.1007/s11060-007-9439-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Accepted: 06/11/2007] [Indexed: 02/06/2023]
Abstract
According to new hypotheses astrocytomas/gliomas either arise from or attract neural stem cells. Biological markers, particularly antigenic markers, have played a significant role for the characterization of these tumour stem cells (TSCc). Because these studies have been performed with single experimental samples mostly from gliomas, we investigated the expression of the stem cell markers CD133/Prominin, Nestin, Sox-2, Musashi-1, CXCR4, Flt-4/VEGFR-3 and CD105/Endoglin in 72 astrocytomas of different WHO-grades and compared it to normal adult human brain. Expression of their mRNA was quantified by quantitative RT-PCR, of their protein by counting immunopositive cells. In contrast to normal brain, tumour samples showed a high variability for the expression of all markers. However, their mean expression was significantly increased in astrocytomas, but this depended on the WHO grade only for CD133, Nestin, Sox-2 and Musashi-1. Confocal microscopy revealed that these markers mostly could be co-stained with glial fibrillary acidic protein, a marker for astoglial cells, but less frequently with the proliferation marker Ki-67/MIB-1. These markers sometimes, but not necessarily could be co-stained with each other in complex patterns. Our results show that most astrocytomas contain considerable portions of cells expressing stem cell markers. It appears that some of these cells originate from tumour genesis (supporting the stem cell hypothesis) while others are attracted by the tumours. Further functional markers are required to differentiate these TSC-types.
Collapse
Affiliation(s)
- Yue-Hui Ma
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, Campus Kiel, Schittenhelmstr. 10, 24105, Kiel, Germany
| | | | | | | | | | | |
Collapse
|
225
|
Galvin KE, Ye H, Wetmore C. Differential gene induction by genetic and ligand-mediated activation of the Sonic hedgehog pathway in neural stem cells. Dev Biol 2007; 308:331-42. [PMID: 17599824 DOI: 10.1016/j.ydbio.2007.05.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 05/21/2007] [Accepted: 05/22/2007] [Indexed: 11/18/2022]
Abstract
Sonic hedgehog (Shh), a secreted morphogen and mitogen, is essential for nervous system development and neural stem cell (NSC) self-renewal. As the intracellular signal transduction of Shh in NSCs is largely unknown, we sought to characterize pathway targets using ligand stimulation and genetic models of activation. NSCs haploinsufficient for Patched (Ptc), a receptor repressive to Shh signaling, showed enhanced proliferation of a magnitude similar to Shh-treated wild-type (Wt) NSCs. Analysis of the Gli zinc-finger transcription factors, primary mediators of Shh activity, demonstrated differential induction between models of pathway activation. Gli1 was significantly induced in Wt NSCs exposed to Shh, whereas Gli2 was elevated and Gli1 expression did not change in Ptc(+/-) NSCs. Other Shh targets (Nmyc, Id factors) were induced under both conditions of pathway activation. Interestingly, Shh-treated Ptc(+/-) NSCs induced expression of Gli1 but failed to increase proliferation, suggesting that the NSCs may have reached a physiologic plateau in proliferative capacity. Thus, our data demonstrate that Ptc(+/-) mice have an expanded progenitor cell niche in vivo and that NSCs maintain a cell-intrinsic increase in basal proliferation in vitro that is sustained by a Gli transduction signature distinct from that of exogenous Shh stimulation. Additionally, Ptc(+/-) NSCs maintain tight control over mitosis and do not further augment proliferation in the presence of mitogenic stimulation.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Proliferation/drug effects
- Cells, Cultured
- DNA Primers/genetics
- Gene Expression Regulation, Developmental/drug effects
- Genes, myc
- Hedgehog Proteins/metabolism
- Hedgehog Proteins/pharmacology
- Hippocampus/cytology
- Hippocampus/metabolism
- Inhibitor of Differentiation Proteins/genetics
- Ligands
- Mice
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Mice, Transgenic
- Models, Genetic
- Multipotent Stem Cells/cytology
- Multipotent Stem Cells/drug effects
- Multipotent Stem Cells/metabolism
- Neurons/cytology
- Neurons/drug effects
- Neurons/metabolism
- Patched Receptors
- Patched-1 Receptor
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Recombinant Proteins/pharmacology
- Signal Transduction
- Transcriptional Activation
Collapse
Affiliation(s)
- Katherine E Galvin
- Program in Molecular Neuroscience, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | |
Collapse
|
226
|
Feng YZ, Shiozawa T, Miyamoto T, Kashima H, Kurai M, Suzuki A, Ying-Song J, Konishi I. Overexpression of hedgehog signaling molecules and its involvement in the proliferation of endometrial carcinoma cells. Clin Cancer Res 2007; 13:1389-98. [PMID: 17332280 DOI: 10.1158/1078-0432.ccr-06-1407] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Research has revealed abnormal activation of the hedgehog pathway in human malignancies. The present study was undertaken to examine the expression and functional involvement of the hedgehog pathway in endometrial tissues. EXPERIMENTAL DESIGN The expression of sonic hedgehog (Shh), patched (Ptch), Smoothened (Smo), and Gli1 was examined in various endometrial tissues and endometrial carcinoma cell lines. The effect of hedgehog signaling on the proliferation of endometrial carcinoma cell lines was also examined. RESULTS The expression of Shh, Ptch, Smo, and Gli1 was very weak in normal endometrium, but was increased in endometrial hyperplasia and carcinoma stepwisely with significant differences. There was no marked difference in the expression of these molecules in carcinomas according to stages and histologic grades. Treatment with cyclopamine, a specific inhibitor of the hedgehog pathway, for endometrial carcinoma Ishikawa and HHUA cells suppressed growth by 56% and 67%, respectively, compared with the control. The addition of recombinant Shh peptide to HHUA cells enhanced their proliferation by 41%. The silencing of Gli1 using small interfering RNA (siGli1) resulted in the growth suppression and down-regulation of Ptch expression. In addition, the cyclopamine/siGli1-induced growth suppression was associated with the down-regulation of cyclins D1 and A and N-myc. No somatic mutations for ptch and smo genes were detected in the endometrial carcinoma cases examined. CONCLUSIONS The abnormal activation of this pathway is involved in the proliferation of endometrial carcinoma cells possibly in an auto-/paracrine fashion, suggesting the possibility of the hedgehog pathway being a novel candidate for molecular targeting.
Collapse
Affiliation(s)
- Yu-Zhen Feng
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, Asahi, Matsumoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
227
|
Perlecan controls neurogenesis in the developing telencephalon. BMC DEVELOPMENTAL BIOLOGY 2007; 7:29. [PMID: 17411441 PMCID: PMC1852307 DOI: 10.1186/1471-213x-7-29] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Accepted: 04/05/2007] [Indexed: 01/17/2023]
Abstract
Background Perlecan is a proteoglycan expressed in the basal lamina of the neuroepithelium during development. Perlecan absence does not impair basal lamina assembly, although in the 55% of the mutants early disruptions of this lamina conducts to exencephaly, impairing brain development. The rest of perlecan-null brains complete its prenatal development, maintain basal lamina continuity interrupted by some isolated ectopias, and are microcephalic. Microcephaly consists of thinner cerebral walls and underdeveloped ganglionic eminences. We have studied the mechanisms that generate brain atrophy in telencephalic areas where basal lamina is intact. Results Brain atrophy in the absence of perlecan started in the ventral forebrain and extended to lateral and dorsal parts of the cortex in the following stages. First, the subpallial forebrain developed poorly in early perlecan-null embryos, because of a reduced cell proliferation: the number of cells in mitosis decreased since the early stages of development. This reduction resulted in a decreased tangential migration of interneurons to the cerebral cortex. Concomitant with the early hypoplasia observed in the medial ganglionic eminences, Sonic Hedgehog signal decreased in the perlecan-null floor plate basal lamina at E12.5. Second, neurogenesis in the pallial neuroepithelium was affected in perlecan deficient embryos. We found reductions of nearly 50% in the number of cells exiting the cell cycle at E12–E13. The labeling index, which was normal at this age, significantly decreased with advancing corticogenesis. Moreover, nestin+ or PCNA+ progenitors increased since E14.5, reaching up to about 150% of the proportion of PCNA+ cells in the wild-type at E17.5. Thus, labeling index reduction together with increased progenitor population, suggests that atrophy is the result of altered cell cycle progression in the cortical progenitors. Accordingly, less neurons populated the cortical plate and subplate of perlecan-null neocortex, as seen with the neuronal markers β-tubulin and Tbr1. Conclusion As a component of the basal lamina, perlecan both maintains this structure and controls the response of the neuroepithelium to growth factors. Less mitotic cells in the early medial ganglionic eminences, and impaired cell cycle progression in the late neocortex, suggests insufficient recruitment and signaling by neurogenic morphogens, such as SHH or FGF2.
Collapse
|
228
|
Eberhart CG. In search of the medulloblast: neural stem cells and embryonal brain tumors. Neurosurg Clin N Am 2007; 18:59-69, viii-ix. [PMID: 17244554 DOI: 10.1016/j.nec.2006.10.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Medulloblastomas have a cellular and molecular phenotype similar in many ways to that of neural stem cells. Indeed, it has long been believed that a medulloblastoma can arise from transformed neural stem cells. Recent analyses of murine transgenic lines has confirmed that cells of the external germinal layer (EGL) can be transformed into a medulloblastoma, generally in association with activation of the Hedgehog signaling pathway. Stem or progenitor cell populations outside the EGL, however, are also likely the cells of origin for a subset of medulloblastomas. Many nonnodular tumors, for example, express markers suggesting that they derive from the ventricular zone germinal layer and show evidence of Wnt pathway activation. Understanding the role of developmental signaling pathways, such as Hedgehog and Wnt, in the initiation and growth of embryonal brain tumors may lead to novel therapies for these highly malignant lesions. In addition, because such pathways are required in neural stem cells, their blockade may prove particularly effective in ablating the stem-like cells within medulloblastomas that are critical for tumor propagation. In support of this concept, inhibition of a third pathway important in stem cells, Notch, seems to deplete the stem-like tumor fraction and block formation of xenografts.
Collapse
Affiliation(s)
- Charles G Eberhart
- Division of Neuropathology, Department of Pathology, Ross Building 558, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| |
Collapse
|
229
|
Guner B, Karlstrom RO. Cloning of zebrafish nkx6.2 and a comprehensive analysis of the conserved transcriptional response to Hedgehog/Gli signaling in the zebrafish neural tube. Gene Expr Patterns 2007; 7:596-605. [PMID: 17307034 PMCID: PMC2043473 DOI: 10.1016/j.modgep.2007.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 01/02/2007] [Accepted: 01/05/2007] [Indexed: 11/30/2022]
Abstract
Sonic Hedgehog (Shh) signaling helps pattern the vertebrate neural tube, in part by regulating the dorsal/ventral expression of a number of homeodomain containing transcription factors. These Hh responsive genes have been divided into two classes, with Class II genes being activated by Hh signaling and Class I genes being repressed by Hh signaling. While the transcriptional response to varying Hh levels is well defined in chick and mouse, it is only partially described in zebrafish, despite the fact that zebrafish has emerged as a powerful genetic system for the study of neural patterning. To better characterize the Hh response in the zebrafish neural tube, we cloned the zebrafish Class II Hh target genes nkx2.9 and nkx6.2. We then analyzed the expression of a number of Class I and Class II Hh responsive genes in wild type, Hh mutant, and Hh over-expressing zebrafish embryos. We show that expression of Class I and Class II genes is highly conserved in the vertebrate neural tube. Further, ventral-most Class II gene expression was completely lost in all Hh pathway mutants analyzed, indicating high levels of Hh signaling are blocked in all of these mutants. In contrast, more dorsally expressed genes were variably affected in different Hh pathway mutants, indicating mid-levels of Hh signaling are differentially affected. This comprehensive expression study provides an important tool for the characterization of Hh signaling in zebrafish and provides a sensitive assay for determining the degree to which newly identified zebrafish mutants affect Hh signaling.
Collapse
MESH Headings
- Animals
- Cloning, Molecular
- DNA, Complementary/genetics
- DNA, Complementary/metabolism
- Embryo, Nonmammalian/cytology
- Embryo, Nonmammalian/metabolism
- Gene Expression Regulation, Developmental
- Hedgehog Proteins/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- In Situ Hybridization
- Neurons/cytology
- Neurons/metabolism
- RNA Probes
- RNA, Messenger/administration & dosage
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/metabolism
Collapse
Affiliation(s)
- Burcu Guner
- Biology Department, University of Massachusetts, Amherst, MA 01003-9297, USA
| | | |
Collapse
|
230
|
Zeng Z, Yuan X, Liu G, Zeng X, Zeng X, Ng H, Chen H, Jiang T, Akasaki Y, Kessey K, Black KL, Yu JS. Manipulation of proliferation and differentiation of human bone marrow-derived neural stem cells in vitro and in vivo. J Neurosci Res 2007; 85:310-20. [PMID: 17131390 DOI: 10.1002/jnr.21131] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Recent evidence has demonstrated that neural stem cells (NSC) can be expanded from a variety of sources, including embryos, fetuses, and adult bone marrow and brain tissue. We have previously reported the generation of adult rat bone marrow-derived cellular spheres that are morphologically and phenotypically similar to neurospheres derived from brain NSC. Here we show that adult human bone marrow-derived neural stem cells (HBM-NSC) are capable of generating spheres that are similar to brain neural-derived neurospheres. Additionally, we sought to promote proliferation and differentiation of HBM-NSC through transduction with nonreplicative recombinant adenovirus encoding the cDNA sequence for Gli, rADV-Gli-1; sonic hedgehog, rADV-Shh; or Nurr1, rADV-Nurr1. Immunocytochemistry and RT-PCR analysis showed that HBM-NSC could be efficiently expanded and differentiated in vitro and that HBM-NSC transduced with rADV-Gli-1 or rADV-Shh dramatically increased NSC time-related proliferation; however, Nurr1 had no effect on proliferation. We also transplanted HBM-NSC into chicken embryos to examine their potential function in vivo. We found that transduction of HBM-NSC with rADV-Gli-1 or rADV-Shh and subsequent transplantation into chicken embryos increased HBM-NSC proliferation, whereas rADV-Nurr1 promoted migration and differentiation in vivo. Our findings suggest that HBM-NSC can be efficiently expanded and differentiated in vitro and in vivo by overexpressing Gli-1, Shh or Nurr1.
Collapse
Affiliation(s)
- Zhaohui Zeng
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Stecca B, Mas C, Clement V, Zbinden M, Correa R, Piguet V, Beermann F, Ruiz i Altaba A. Melanomas require HEDGEHOG-GLI signaling regulated by interactions between GLI1 and the RAS-MEK/AKT pathways. Proc Natl Acad Sci U S A 2007; 104:5895-900. [PMID: 17392427 PMCID: PMC1838820 DOI: 10.1073/pnas.0700776104] [Citation(s) in RCA: 422] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Melanoma is one of the most aggressive cancers, and its incidence is increasing. These tumors derive from the melanocyte lineage and remain incurable after metastasis. Here we report that SONIC HEDGEHOG (SHH)-GLI signaling is active in the matrix of human hair follicles, and that it is required for the normal proliferation of human melanocytes in culture. SHH-GLI signaling also regulates the proliferation and survival of human melanomas: the growth, recurrence, and metastasis of melanoma xenografts in mice are prevented by local or systemic interference of HH-GLI function. Moreover, we show that oncogenic RAS-induced melanomas in transgenic mice express Gli1 and require Hh-Gli signaling in vitro and in vivo. Finally, we provide evidence that endogenous RAS-MEK and AKT signaling regulate the nuclear localization and transcriptional activity of GLI1 in melanoma and other cancer cells. Our data uncover an unsuspected role of HH-GLI signaling in melanocytes and melanomas, demonstrate a role for this pathway in RAS-induced tumors, suggest a general integration of the RAS/AKT and HH-GLI pathways, and open a therapeutic approach for human melanomas.
Collapse
Affiliation(s)
- Barbara Stecca
- Department of Genetic Medicine and Development, University of Geneva School of Medicine, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Christophe Mas
- Department of Genetic Medicine and Development, University of Geneva School of Medicine, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Virginie Clement
- Department of Genetic Medicine and Development, University of Geneva School of Medicine, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Marie Zbinden
- Department of Genetic Medicine and Development, University of Geneva School of Medicine, 1 Rue Michel Servet, 1211 Geneva, Switzerland
| | - Rafael Correa
- Department of Dermatology and Venereology, University Hospital of Geneva, 24 Rue Micheli du Crest, 1211 Geneva, Switzerland; and
| | - Vincent Piguet
- Department of Dermatology and Venereology, University Hospital of Geneva, 24 Rue Micheli du Crest, 1211 Geneva, Switzerland; and
| | - Friedrich Beermann
- Swiss Institute for Experimental Cancer Research, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | - Ariel Ruiz i Altaba
- Department of Genetic Medicine and Development, University of Geneva School of Medicine, 1 Rue Michel Servet, 1211 Geneva, Switzerland
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
232
|
Quiñones-Hinojosa A, Chaichana K. The human subventricular zone: a source of new cells and a potential source of brain tumors. Exp Neurol 2007; 205:313-24. [PMID: 17459377 DOI: 10.1016/j.expneurol.2007.03.016] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2006] [Revised: 02/23/2007] [Accepted: 03/13/2007] [Indexed: 02/04/2023]
Abstract
The mammalian brain has been perceived as a quiescent organ incapable of postnatal neurogenesis for many years. Most recently, several studies have demonstrated that the adult mammalian brain is indeed capable of neurogenesis and that the process is primarily confined to the subventricular zone (SVZ) of the forebrain and the subgranular zone (SGZ) of the hippocampus. Of these regions, the SVZ is the largest niche of neurogenesis in the adult mammalian brain. Within this niche resides a subpopulation of astrocytes with stem cell-like features of self-renewal and multipotentiality. Interestingly, there is also a subpopulation of cells within brain tumors that possess these same characteristics. Based on these findings, the emerging hypothesis is that brain tumor stem cells may be derived from neural stem cells and that both of these populations may originate from the SVZ. This possible connection stresses the importance of studying and understanding the role that the human SVZ plays in not only harboring neural and brain tumor stem cells, but how this microenvironment may support both neurogenesis and tumorigenesis. Furthermore, the obvious differences in the SVZ between humans and other animals make it important to understand the human model when studying human disease. Such an understanding may lead to novel therapeutic strategies for both neurodegenerative diseases and currently intractable brain tumors.
Collapse
Affiliation(s)
- Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
233
|
Ehtesham M, Sarangi A, Valadez JG, Chanthaphaychith S, Becher MW, Abel TW, Thompson RC, Cooper MK. Ligand-dependent activation of the hedgehog pathway in glioma progenitor cells. Oncogene 2007; 26:5752-61. [PMID: 17353902 DOI: 10.1038/sj.onc.1210359] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The hedgehog (Hh) signaling pathway regulates progenitor cells during embryogenesis and tumorigenesis in multiple organ systems. We have investigated the activity of this pathway in adult gliomas, and demonstrate that the Hh pathway is operational and activated within grade II and III gliomas, but not grade IV de novo glioblastoma multiforme. Furthermore, our studies reveal that pathway activity and responsiveness is confined to progenitor cells within these tumors. Additionally, we demonstrate that Hh signaling in glioma progenitor cells is ligand-dependent and provide evidence documenting the in vivo source of Sonic hedgehog protein. These findings suggest a regulatory role for the Hh pathway in progenitor cells within grade II and III gliomas, and the potential clinical utility of monitoring and targeting this pathway in these primary brain tumors.
Collapse
Affiliation(s)
- M Ehtesham
- Department of Neurological Surgery, Vanderbilt Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
234
|
Mistretta CM, Liu HX. Development of fungiform papillae: patterned lingual gustatory organs. ACTA ACUST UNITED AC 2007; 69:199-208. [PMID: 17287575 DOI: 10.1679/aohc.69.199] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The fungiform papilla is a gustatory organ that provides a specific tissue residence for taste buds on the anterior tongue. Thus, during development there must be a progressive differentiation to acquire papilla epithelium, then taste cell progenitor epithelium, and finally taste cells within the papilla apex. Arranged in rows, the patterned distribution of fungiform papillae requires molecular regulation not only to induce papillae, but also to suppress papilla formation in the between-papilla tissue. Intact sensory innervation is not required to initiate papilla development or pattern. However, members of several molecular families have now been identified with specific localization in developing papillae. These may participate in papilla development and pattern formation, and subsequently in taste progenitor and taste cell differentiation. This review focuses on development of fungiform papillae in embryonic rat and mouse. Basic morphology, cell biology and molecular phenotypes of developing papillae are reviewed. Regulatory roles for molecules in several families are presented, and a broad schema is proposed for progressive epithelial differentiation to form taste cell progenitors in parallel with the temporal course, and participation of lingual sensory innervation.
Collapse
Affiliation(s)
- Charlotte M Mistretta
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI 48109-1078, USA.
| | | |
Collapse
|
235
|
Abstract
PURPOSE Recent discoveries highlight the importance of the hedgehog signaling pathway in prostate growth regulation. We reviewed the role of hedgehog signaling in prostate development, adult prostate homeostasis and prostate cancer. MATERIALS AND METHODS A comprehensive review of all relevant literature was done. RESULTS Epithelial expression of hedgehog ligand during prostate development exerts autocrine and paracrine signaling activities that regulate growth and differentiation. Hedgehog signaling also occurs in the adult human prostate but to our knowledge the influence on epithelial proliferation and/or differentiation is unknown. Robust hedgehog signaling occurs frequently in prostate cancer, and autocrine and paracrine signaling have been shown to accelerate the growth of xenograft tumors. Autocrine signaling has been implicated in stimulating stem/progenitor cells and increased hedgehog pathway activity may be a characteristic of advanced, androgen independent cancer. The plant alkaloid cyclopamine is a specific chemical inhibitor of hedgehog signaling that produced sustained regression of established xenograft tumors. CONCLUSIONS Hedgehog signaling has an important role in prostate development and it appears to be a characteristic feature of prostate cancer. It stimulates tumor growth and may exert a specific role in the proliferation of tumor stem cells. The development of hedgehog inhibitors based on the action of cyclopamine holds promise for novel treatments to slow or arrest tumor growth.
Collapse
Affiliation(s)
- Aubie Shaw
- McArdle Laboratory for Cancer Research and Department of Surgery, University of Wisconsin, Madison, Wisconsin 53792, USA
| | | |
Collapse
|
236
|
Bar EE, Chaudhry A, Farah MH, Eberhart CG. Hedgehog signaling promotes medulloblastoma survival via Bc/II. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:347-55. [PMID: 17200206 PMCID: PMC1762704 DOI: 10.2353/ajpath.2007.060066] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activation of the Hedgehog (Hh) pathway has been identified in several cancers, including medulloblastoma, but the mechanisms by which this pathway affects tumor survival and growth are incompletely understood. We investigated whether Hedgehog might promote survival of medulloblastoma cells via up-regulation of BclII. We found that mRNA levels of the Hedgehog pathway effector Gli1 were significantly associated with BclII expression in medulloblastoma and that Gli1 and BclII are both present in regions of decreased apoptosis in nodular medulloblastoma. Transient overexpression of Gli1 and Gli2 in medulloblastoma cultures induced a BclII transcriptional reporter and increased BclII protein levels, whereas stable overexpression of Gli1 was associated with increased BclII mRNA. The Hedgehog antagonist cyclopamine blocked expression of the Hh pathway targets PTCH1 and Gli1, lowered BclII levels, and increased apoptosis in DAOY and UW228 medulloblastoma cells. Apoptotic induction caused by cyclopamine could be rescued in part by enforced expression of Gli1 or BclII. Hh pathway blockade also sensitized medulloblastoma to the effects of the proapoptotic agent lovastatin. These data demonstrate that BclII is an important mediator of Hh activity in medulloblastoma and suggest new strategies for combined chemotherapeutic regimens.
Collapse
Affiliation(s)
- Eli E Bar
- Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Ave., Ross Building 558, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
237
|
Abstract
Several key signalling pathways, such as Hedgehog, Notch, Wnt and BMP-TGFbeta-Activin (bone morphogenetic protein-transforming growth factor-beta-Activin), are involved in most processes essential to the proper development of an embryo. It is also becoming increasingly clear that these pathways can have a crucial role in tumorigenesis when reactivated in adult tissues through sporadic mutations or other mechanisms. We will focus here on the Hedgehog pathway, which is abnormally activated in most basal cell carcinomas, and discuss potential therapeutic opportunities offered by the progress made in understanding this signalling pathway.
Collapse
Affiliation(s)
- Lee L Rubin
- Harvard Stem Cell Institute, Biolabs, Room 150, 16 Divinity Avenue, Cambridge, Massachusetts 02138, USA.
| | | |
Collapse
|
238
|
Clark PA, Treisman DM, Ebben J, Kuo JS. Developmental signaling pathways in brain tumor-derived stem-like cells. Dev Dyn 2007; 236:3297-308. [DOI: 10.1002/dvdy.21381] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
239
|
Lathia JD, Rao MS, Mattson MP, ffrench-Constant C. The microenvironment of the embryonic neural stem cell: Lessons from adult niches? Dev Dyn 2007; 236:3267-82. [DOI: 10.1002/dvdy.21319] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
240
|
Ingham PW, Placzek M. Orchestrating ontogenesis: variations on a theme by sonic hedgehog. Nat Rev Genet 2006; 7:841-50. [PMID: 17047684 DOI: 10.1038/nrg1969] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Embryonic development is an emergent process in which increasing complexity is generated by sequential cellular interactions. Recently, it has become clear that such interactions are mediated by just a few families of signalling molecules; but how does this limited repertoire elicit the diversity of form that is characteristic of multicellular organisms? Here we review the various ways in which a member of one such family, the sonic hedgehog (SHH) protein, is deployed during embryonic development. These examples of SHH function provide paradigms for inductive interactions that should help to inform attempts to recapitulate cellular programming and organogenesis in vitro.
Collapse
Affiliation(s)
- Philip W Ingham
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, 138673, Singapore.
| | | |
Collapse
|
241
|
Mao J, Ligon KL, Rakhlin EY, Thayer SP, Bronson RT, Rowitch D, McMahon AP. A novel somatic mouse model to survey tumorigenic potential applied to the Hedgehog pathway. Cancer Res 2006; 66:10171-8. [PMID: 17047082 PMCID: PMC3806052 DOI: 10.1158/0008-5472.can-06-0657] [Citation(s) in RCA: 225] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We report a novel mouse model for the generation of sporadic tumors and show the efficiency of this approach by surveying Hedgehog (Hh)-related tumors. Up-regulation of the Hh pathway is achieved by conditionally regulated expression of an activated allele of Smoothened (R26-SmoM2) using either sporadic leakage or global postnatal induction of a ubiquitously expressed inducible Cre transgene (CAGGS-CreER). Following postnatal tamoxifen induction, CAGGS-CreER; R26-SmoM2 mice developed tumors with short latency and high penetrance. All mice exhibited rhabdomyosarcoma and basal cell carcinoma; 40% also developed medulloblastoma. In addition, mice showed a novel pancreatic lesion resembling low-grade mucinous cystic neoplasms in humans. In contrast, widespread activation of SmoM2 in the postnatal prostate epithelium results in no detectable morphologic outcome in 12-month-old mice. Comparison of gene expression profiles among diverse tumors identified several signature genes, including components of platelet-derived growth factor and insulin-like growth factor pathways, which may provide a common mechanistic link to the Hh-related malignancies. This experimental model provides a robust tool for exploring the process of Hh-dependent tumorigenesis and the treatment of such tumors. More generally, this approach provides a genetic platform for identifying tumorigenic potential in putative oncogenes and tumor suppressors and for more effective modeling of sporadic cancers in mice.
Collapse
Affiliation(s)
- Junhao Mao
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts
| | - Keith L. Ligon
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical Schoo, Boston, Massachusetts
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Elena Y. Rakhlin
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sarah P. Thayer
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Roderick T. Bronson
- Department of Biomedical Sciences, Tufts University Veterinary School, North Grafton, Massachusetts
| | - David Rowitch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical Schoo, Boston, Massachusetts
| | - Andrew P. McMahon
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
242
|
Kise Y, Takenaka K, Tezuka T, Yamamoto T, Miki H. Fused kinase is stabilized by Cdc37/Hsp90 and enhances Gli protein levels. Biochem Biophys Res Commun 2006; 351:78-84. [PMID: 17054904 DOI: 10.1016/j.bbrc.2006.10.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2006] [Accepted: 10/01/2006] [Indexed: 12/21/2022]
Abstract
Serine/threonine kinase Fused (Fu) is an essential component of Hedgehog (Hh) signaling in Drosophila, but the biochemical functions of Fu remain unclear. Here, we have investigated proteins co-precipitated with mammalian Fu and identified a kinase-specific chaperone complex, Cdc37/Hsp90, as a novel-binding partner of Fu. Inhibition of Hsp90 function by geldanamycin (GA) induces rapid degradation of Fu through a ubiquitin-proteasome pathway. We next show that co-expression of Fu with transcription factors Gli1 and Gli2 significantly increases their protein levels and luciferase reporter activities, which are blocked by GA. These increases can be ascribed to Fu-mediated stabilization of Gli because co-expression of Fu prolongs half-life of Gli1 and reduces polyubiquitination of Gli1. Finally, we show that GA inhibits proliferation of PC3, a Hh signaling-activated prostate cancer cell line. This growth inhibition is partially rescued by expression of ectopic Gli1, suggesting that Fu may contribute to enhance Hh signaling activity in cancer cells.
Collapse
Affiliation(s)
- Yoshiaki Kise
- Division of Cancer Genomics, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | |
Collapse
|
243
|
Bertrand N, Dahmane N. Sonic hedgehog signaling in forebrain development and its interactions with pathways that modify its effects. Trends Cell Biol 2006; 16:597-605. [PMID: 17030124 DOI: 10.1016/j.tcb.2006.09.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2006] [Revised: 09/05/2006] [Accepted: 09/25/2006] [Indexed: 11/21/2022]
Abstract
During the development of the nervous system and other organs in the embryo, a limited set of master signaling pathways are used repeatedly for induction, patterning and growth. Among these, the Sonic hedgehog (Shh) pathway is crucial for the development of many structures in the brain. How the context-specific interplay between these various signaling pathways produces distinct temporal and spatial outcomes is not clear. Resolving this problem is a major goal in the study of cell and organ development. Here, we focus on signaling events during dorso-ventral patterning of the embryonic forebrain in vertebrates. In particular, we discuss the role of the Shh pathway in this process and on its interactions with the FGF, retinoic acid and Nodal pathways and other information cascades that modify its effects.
Collapse
Affiliation(s)
- Nicolas Bertrand
- IBDML, UMR 6216, Campus de Luminy, Université de la Méditerranée, 13288 Marseille cedex 09, France
| | | |
Collapse
|
244
|
The effects of PACAP on neural cell proliferation. ACTA ACUST UNITED AC 2006; 137:50-7. [PMID: 17011642 DOI: 10.1016/j.regpep.2006.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 03/20/2006] [Accepted: 03/30/2006] [Indexed: 01/25/2023]
Abstract
PACAP and its receptors are expressed in growth zones of the brain. By stimulating PAC(1)-receptors PACAP can enhance, as well as reduce, the proliferation rate in a cell-type dependent manner. PACAP can enhance the proliferation rate by activating phospholipase C and protein kinase C, although other signal transduction pathways may also be responsible. PACAP can suppress proliferation by inhibiting protein complexes of the cyclins D and E with the cyclin-dependent kinases 4/6 and 2, respectively, which are necessary for entry into the cell cycle. PACAP seems to exert these inhibitory effects by acting via the Sonic hedgehog glycoprotein and the small GTPase RhoA. Also, the activation of a cyclin-dependent kinase inhibitor has been suggested. The signal transduction pathways mediating the effects of PACAP on proliferation are discussed.
Collapse
|
245
|
Navarro-Quiroga I, Hernandez-Valdes M, Lin SL, Naegele JR. Postnatal cellular contributions of the hippocampus subventricular zone to the dentate gyrus, corpus callosum, fimbria, and cerebral cortex. J Comp Neurol 2006; 497:833-45. [PMID: 16786555 DOI: 10.1002/cne.21037] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The rodent dentate gyrus (DG) is formed in the embryo when progenitor cells migrate from the dentate neuroepithelium to establish a germinal zone in the hilus and a secondary germinal matrix, near the fimbria, called the hippocampal subventricular zone (HSVZ). The developmental plasticity of progenitors within the HSVZ is not well understood. To delineate the migratory routes and fates of progenitors within this zone, we injected a replication-incompetent retrovirus, encoding the enhanced green fluorescent protein (EGFP), into the HSVZ of postnatal day 5 (P5) mice. Between P6 and P45, retrovirally-infected EGFP(+) of progenitors migrated into the DG, established a reservoir of progenitor cells, and differentiated into neurons and glia. By P6-7, EGFP(+) cells were observed migrating into the DG. Subsets of these EGFP(+) cells expressed Sox2 and Musashi-1, characteristic of neural stem cells. By P10, EGFP(+) cells assumed positions within the DG and expressed immature neuronal markers. By P20, many EGFP(+) cells expressed the homeobox prospero-like protein Prox1, an early and specific granule cell marker in the CNS, and extended mossy fiber projections into the CA3. A subset of non-neuronal EGFP(+) cells in the dentate gyrus acquired the morphology of astrocytes. Another subset included EGFP(+)/RIP(+) oligodendrocytes that migrated into the fimbria, corpus callosum, and cerebral cortex. Retroviral injections on P15 labeled very few cells, suggesting depletion of HSVZ progenitors by this age. These findings suggest that the early postnatal HSVZ progenitors are multipotent and migratory, and contribute to both dentate gyrus neurogenesis as well as forebrain gliogenesis.
Collapse
|
246
|
Kasper M, Schnidar H, Neill GW, Hanneder M, Klingler S, Blaas L, Schmid C, Hauser-Kronberger C, Regl G, Philpott MP, Aberger F. Selective modulation of Hedgehog/GLI target gene expression by epidermal growth factor signaling in human keratinocytes. Mol Cell Biol 2006; 26:6283-98. [PMID: 16880536 PMCID: PMC1592816 DOI: 10.1128/mcb.02317-05] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 04/27/2006] [Accepted: 06/02/2006] [Indexed: 11/20/2022] Open
Abstract
Hedgehog (HH)/GLI signaling plays a critical role in epidermal development and basal cell carcinoma. Here, we provide evidence that epidermal growth factor receptor (EGFR) signaling modulates the target gene expression profile of GLI transcription factors in epidermal cells. Using expression profiling and quantitative reverse transcriptase PCR, we identified a set of 19 genes whose transcription is synergistically induced by GLI1 and parallel EGF treatment. Promoter studies of a subset of GLI/EGF-regulated genes, including the genes encoding interleukin-1 antagonist IL1R2, Jagged 2, cyclin D1, S100A7, and S100A9, suggest convergence of EGFR and HH/GLI signaling at the level of promoters of selected direct GLI target genes. Inhibition of EGFR and MEK/ERK but not of phosphatidylinositol 3-kinase/AKT abrogated synergistic activation of GLI/EGF target genes, showing that EGFR can signal via RAF/MEK/ERK to cooperate with GLI proteins in selective target gene regulation. Coexpression of the GLI/EGF target IL1R2, EGFR, and activated ERK1/2 in human anagen hair follicles argues for a cooperative role of EGFR and HH/GLI signaling in specifying the fate of outer root sheath (ORS) cells. We also show that EGF treatment neutralizes GLI-mediated induction of epidermal stem cell marker expression and provide evidence that EGFR signaling is essential for GLI-induced cell cycle progression in epidermal cells. The results suggest that EGFR signaling modulates GLI target gene profiles which may play an important regulatory role in ORS specification, hair growth, and possibly cancer.
Collapse
Affiliation(s)
- Maria Kasper
- Department of Molecular Biology, University of Salzburg, Hellbrunnerstrasse 34, A-5020 Salzburg, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Abstract
The hedgehog pathway is a major regulator of embryonic development, and mutations that decrease its activity are known to be associated with severe defects in nervous system development. Recent evidence suggests hedgehog continues to function in adult tissue, normal as well as diseased, by regulating both cell proliferation and the production of growth and angiogenic factors. In the adult nervous system, this dual ability is especially important in regulating the behavior of neural stem and progenitor cells. This review summarizes information connecting hedgehog signaling and neural diseases, including neurodegenerative disorders and brain tumors, particularly medulloblastoma. We also describe the discovery and utility of small molecule agonists and antagonists of this pathway and their potential as novel types of therapeutics.
Collapse
|
248
|
Rothschild G, Zhao X, Iavarone A, Lasorella A. E Proteins and Id2 converge on p57Kip2 to regulate cell cycle in neural cells. Mol Cell Biol 2006; 26:4351-61. [PMID: 16705184 PMCID: PMC1489106 DOI: 10.1128/mcb.01743-05] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
A precise balance between proliferation and differentiation must be maintained during neural development to obtain the correct proportion of differentiated cell types in the adult nervous system. The basic helix-loop-helix (bHLH) transcription factors known as E proteins and their natural inhibitors, the Id proteins, control the timing of differentiation and terminal exit from the cell cycle. Here we show that progression into S phase of human neuroblastoma cells is prevented by E proteins and promoted by Id2. Cyclin-dependent kinase inhibitors (CKI) have been identified as key effectors of cell cycle arrest in differentiating cells. However, p57Kip2 is the only CKI that is absolutely required for normal development. Through the use of global gene expression analysis in neuroblastoma cells engineered to acutely express the E protein E47 and Id2, we find that p57Kip2 is a target of E47. Consistent with the role of Id proteins, Id2 prevents activation of p57Kip2 expression, and the retinoblastoma tumor suppressor protein, a known Id2 inhibitor, counters this activity. The strong E47-mediated inhibition of entry into S phase is entirely reversed in cells in which expression of p57Kip2 is silenced by RNA interference. During brain development, expression of p57Kip2 is opposite that of Id2. Our findings identify p57Kip2 as a functionally relevant target recruited by bHLH transcription factors to induce cell cycle arrest in developing neuroblasts and suggest that deregulated expression of Id proteins may be an epigenetic mechanism to silence expression of this CKI in neural tumors.
Collapse
Affiliation(s)
- Gerson Rothschild
- Institute for Cancer Genetics, 1150 St. Nicholas Ave., Columbia University, New York, NY 10032, USA
| | | | | | | |
Collapse
|
249
|
Zhang W, Yi MJ, Chen X, Cole F, Krauss RS, Kang JS. Cortical thinning and hydrocephalus in mice lacking the immunoglobulin superfamily member CDO. Mol Cell Biol 2006; 26:3764-72. [PMID: 16648472 PMCID: PMC1489002 DOI: 10.1128/mcb.26.10.3764-3772.2006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
CDO is a cell surface immunoglobulin superfamily member that positively regulates myogenic differentiation in vitro and in vivo and signals to posttranslationally activate myogenic basic helix-loop-helix (bHLH) transcription factors. The Cdo gene is also expressed in the dorsal aspect and midline structures of the developing central nervous system, and mice lacking CDO on the C57BL/6 background display holoprosencephaly with approximately 80% penetrance, resulting in perinatal lethality. We report here that a fraction of Cdo-/- mice from this background have additional defects in brain development, including hydrocephalus and cortical thinning. Primary neural progenitor cultures from E14.5 Cdo-/- mutants display reduced proliferation, which may underlie the thinning. The cortical preplate and cortices of mutant animals also show reduced staining for beta-tubulin III, indicating defective neuronal differentiation. CDO levels are strongly increased in cultured C17.2 neuronal precursor cells stimulated to differentiate; modulation of CDO levels in these cells by overexpression or interfering RNA approaches enhances or diminishes differentiation, respectively. Cotransfection of CDO enhances the activity of the neurogenic bHLH factor, neurogenin1, in reporter assays and enhances heterodimerization of neurogenin1 and E47. These results indicate that CDO promotes neuronal differentiation and support the hypothesis that CDO coordinates differentiation of multiple cell lineages by regulating the activity of tissue-specific bHLH factors.
Collapse
Affiliation(s)
- Wei Zhang
- Brookdale Department of Molecular, Cell, and Developmental Biology, Box 1020, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | |
Collapse
|
250
|
Abstract
Both stem cells and cancer cells are thought to be capable of unlimited proliferation. Moreover, many tumours and cancer cell lines express stem cell markers, including adenosine triphosphate (ATP)-binding cassette transporters, by which the cells pump out specific fluorescent dyes as well as anti-cancer drugs, suggesting either that cancer cells resemble stem cells or that cancers contain stem-like cells. Using the common characteristics of brain tumour cells and neural stem cells, several research groups have succeeded in identifying stem-like cells (cancer stem-like cells) in brain tumours and brain cancer cell lines. The purified cancer stem-like cells, but not the other cancer cells, self-renew and form tumours when transplanted in vivo. Thus, cancer stem-like cells in brain tumours might be a crucial target for anti-brain tumour therapy.
Collapse
Affiliation(s)
- Toru Kondo
- Centre for Brain Repair, University of Cambridge, Cambridge CB2 2PY, UK.
| |
Collapse
|