Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Dev Biol ; 397(2): 191-202, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25448692

ABSTRACT

Ptch1 and Ptch2 are highly conserved vertebrate homologs of Drosophila ptc, the receptor of the Hedgehog (Hh) signaling pathway. The vertebrate Ptch1 gene encodes a potent tumor suppressor and is well established for its role in embryonic development. In contrast, Ptch2 is poorly characterized and dispensable for embryogenesis. In flies and mice, ptc/Ptch1 controls Hh signaling through the regulation of Smoothened (Smo). In addition, Hh pathway activation also up-regulates ptc/Ptch1 expression to restrict the diffusion of the ligand. Recent studies have implicated Ptch2 in this ligand dependent antagonism, however whether Ptch2 encodes a functional Shh receptor remains unclear. In this report, we demonstrate that Ptch2 is a functional Shh receptor, which regulates Smo localization and activity in vitro. We also show that Ptch1 and Ptch2 are co-expressed in the developing mouse limb bud and loss of Ptch2 exacerbates the outgrowth defect in the limb-specific Ptch1 knockout mutants, demonstrating that Ptch1 and Ptch2 co-operate in regulating cellular responses to Shh in vivo.


Subject(s)
Extremities/embryology , Morphogenesis/physiology , Receptors, Cell Surface/metabolism , Animals , Blotting, Western , Cell Line , Hedgehog Proteins/metabolism , In Situ Hybridization , Mice , Mice, Knockout , Morphogenesis/genetics , Patched Receptors , Patched-1 Receptor , Patched-2 Receptor , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor
2.
Development ; 139(22): 4152-61, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23034632

ABSTRACT

Abnormal activation of Hedgehog (Hh) signaling leads to basal cell carcinoma (BCC) of the skin, the most common human cancer. Gli2, the major transcriptional activator of Hh signaling, is essential for hair follicle development and its overexpression in epidermis induces BCC formation and maintains tumor growth. Despite its importance in skin development and tumorigenesis, little is known about the molecular regulation of Gli2. Sufu and Kif7 are two evolutionarily conserved regulators of Gli transcription factors. Here, we show that Sufu and Kif7 regulate Gli2 through distinct mechanisms in keratinocytes. Sufu restricts the activity of Gli2 through cytoplasmic sequestration. Kif7 possesses Sufu-dependent and -independent regulatory functions in Hh signaling: while it promotes Hh pathway activity through the dissociation of Sufu-Gli2 complex, it also contributes to the repression of Hh target genes in the absence of Sufu. Deletion of both Sufu and Kif7 in embryonic skin leads to complete loss of follicular fate. Importantly, although inactivation of Sufu or Kif7 alone in adult epidermis cannot promote BCC formation, their simultaneous deletion induces BCC. These studies establish Sufu and Kif7 as crucial components in the regulation of Gli2 localization and activity, and illustrate their overlapping functions in skin development and tumor suppression.


Subject(s)
Carcinoma, Basal Cell/metabolism , Keratinocytes/metabolism , Kinesins/metabolism , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Skin Neoplasms/metabolism , Skin/embryology , Animals , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Cell Proliferation , Cell Transformation, Neoplastic , Cytoplasm , Hair Follicle/embryology , Hedgehog Proteins , Kinesins/deficiency , Kinesins/genetics , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/biosynthesis , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics , Signal Transduction , Skin Neoplasms/pathology , Zinc Finger Protein Gli2
3.
Blood ; 116(4): 653-60, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20339091

ABSTRACT

Despite the clear importance of Hedgehog (Hh) signaling in blood vascular development as shown by genetic analysis, its mechanism of action is still uncertain. To better understand the role of Hh in vascular development, we further characterized its roles in vascular development in mouse embryos and examined its interaction with vascular endothelial growth factor (VEGF), a well-known signaling pathway essential to blood vascular development. We found that VEGF expression in the mouse embryo depended on Hh signaling, and by using genetic rescue approaches, we demonstrated that the role of Hh both in endothelial tube formation and Notch-dependent arterial identity was solely dependent on its regulation of VEGF. In contrast, overactivation of the Hh pathway through deletion of Patched1 (Ptch1), a negative regulator of Hh signaling, resulted in reduced vascular density and increased Delta-like ligand 4 expression. The Ptch1 phenotype was independent of VEGF pathway dysregulation and was not rescued when Delta-like ligand 4 levels were restored to normal. These findings establish that Hh uses both VEGF- and Notch-dependent and -independent mechanisms to pattern specific events in early blood vascular development.


Subject(s)
Hedgehog Proteins/physiology , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor A/physiology , Animals , Blood Vessels/embryology , Blood Vessels/metabolism , Body Patterning/genetics , Embryo, Mammalian , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Mice, Transgenic , Models, Biological , Patched Receptors , Patched-1 Receptor , Pregnancy , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Receptors, Notch/genetics , Receptors, Notch/physiology , Signal Transduction/genetics , Tissue Distribution , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 298(1): L45-56, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19897741

ABSTRACT

Distal lung development occurs through coordinated induction of myofibroblasts, epithelial cells, and capillaries. Lunatic Fringe (Lfng) is a beta(1-3) N-acetylglucosamine transferase that modifies Notch receptors to facilitate their activation by Delta-like (Dll1/4) ligands. Lfng is expressed in the distal lung during saccular development, and deletion of this gene impairs myofibroblast differentiation and alveogenesis in this context. A similar defect was observed in Notch2(beta-geo/+)Notch3(beta-geo/beta-geo) compound mutant mice but not in Notch2(beta-geo/+) or Notch3(beta-geo/beta-geo) single mutants. Finally, to directly test for the role of Notch signaling in myofibroblast differentiation in vivo, we used ROSA26-rtTA(/+);tetO-CRE(/+);RBPJkappa(flox/flox) inducible mutant mice to show that disruption of canonical Notch signaling during late embryonic development prevents induction of smooth muscle actin in mesenchymal cells of the distal lung. In sum, these results demonstrate that Lfng functions to enhance Notch signaling in myofibroblast precursor cells and thereby to coordinate differentiation and mobilization of myofibroblasts required for alveolar septation.


Subject(s)
Glycosyltransferases/metabolism , Organogenesis , Pulmonary Alveoli/embryology , Receptors, Notch/metabolism , Signal Transduction , Alleles , Animals , Cell Differentiation , Collagen/metabolism , Elastin/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Genome/genetics , Immunohistochemistry , Ligands , Mice , Mice, Mutant Strains , Mutation/genetics , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Pulmonary Alveoli/abnormalities , Pulmonary Alveoli/pathology , Stem Cells/metabolism
5.
Mol Cell Biol ; 26(17): 6609-22, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16914743

ABSTRACT

Hedgehog (Hh) signaling plays pivotal roles in tissue patterning and development in Drosophila melanogaster and vertebrates. The Patched1 (Ptc1) gene, encoding the Hh receptor, is mutated in nevoid basal cell carcinoma syndrome, a human genetic disorder associated with developmental abnormalities and increased incidences of basal cell carcinoma (BCC) and medulloblastoma (MB). Ptc1 mutations also occur in sporadic forms of BCC and MB. Mutational studies with mice have verified that Ptc1 is a tumor suppressor. We previously identified a second mammalian Patched gene, Ptc2, and demonstrated its distinct expression pattern during embryogenesis, suggesting a unique role in development. Most notably, Ptc2 is expressed in an overlapping pattern with Shh in the epidermal compartment of developing hair follicles and is highly expressed in the developing limb bud, cerebellum, and testis. Here, we describe the generation and phenotypic analysis of Ptc2(tm1/tm1) mice. Our molecular analysis suggests that Ptc2(tm1) likely represents a hypomorphic allele. Despite the dynamic expression of Ptc2 during embryogenesis, Ptc2(tm1/tm1) mice are viable, fertile, and apparently normal. Interestingly, adult Ptc2(tm1/tm1) male animals develop skin lesions consisting of alopecia, ulceration, and epidermal hyperplasia. While functional compensation by Ptc1 might account for the lack of a strong mutant phenotype in Ptc2-deficient mice, our results suggest that normal Ptc2 function is required for adult skin homeostasis.


Subject(s)
Alopecia/pathology , Fetal Viability , Gene Targeting , Hair Follicle/pathology , Mutation/genetics , Receptors, Cell Surface/metabolism , Animals , Cells, Cultured , Cerebellum/cytology , Cerebellum/embryology , Embryo, Mammalian/cytology , Embryo, Mammalian/pathology , Extremities/embryology , Hair Follicle/cytology , Hair Follicle/embryology , Hedgehog Proteins , Hyperplasia , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Patched Receptors , Patched-1 Receptor , Patched-2 Receptor , Phenotype , Receptors, Cell Surface/deficiency , Signal Transduction , Testis/cytology , Testis/embryology , Trans-Activators/metabolism , Up-Regulation/genetics , Zinc Finger Protein GLI1
6.
J Invest Dermatol ; 134(7): 1981-1990, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24492243

ABSTRACT

The Patched 1 (Ptch1) receptor has a pivotal role in inhibiting the activity of the Hedgehog (Hh) pathway and is therefore critical in preventing the onset of many human developmental disorders and tumor formation. However, the functional role of the mammalian Ptch2 paralogue remains elusive, particularly the extent to which it contributes to regulating the spatial and temporal activity of Hh signaling. Here we demonstrate in three independent mouse models of epidermal development that in vivo ablation of both Ptch receptors results in a more severe phenotype than loss of Ptch1 alone. Our studies indicate that concomitant loss of Ptch1 and Ptch2 activity inhibits epidermal lineage specification and differentiation. These results reveal that repression of Hh signaling through a dynamic Ptch regulatory network is a crucial event in lineage fate determination in the skin. In general, our findings implicate Ptch receptor redundancy as a key issue in elucidating the cellular origin of Hh-induced tumors.


Subject(s)
Epidermal Cells , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Skin Neoplasms , Animals , Animals, Outbred Strains , Cell Differentiation/physiology , Cell Lineage/physiology , Epidermis/embryology , Epidermis/metabolism , Female , Hair Follicle/cytology , Hair Follicle/embryology , Hair Follicle/metabolism , Hedgehog Proteins/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, 129 Strain , Mice, SCID , Patched Receptors , Patched-1 Receptor , Patched-2 Receptor , Pregnancy , Receptors, Cell Surface/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Transplantation
7.
PLoS One ; 4(10): e7313, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19809516

ABSTRACT

Truncating GLI3 mutations in Pallister-Hall Syndrome with renal malformation suggests a requirement for Hedgehog signaling during renal development. HH-dependent signaling increases levels of GLI transcriptional activators and decreases processing of GLI3 to a shorter transcriptional repressor. Previously, we showed that Shh-deficiency interrupts early inductive events during renal development in a manner dependent on GLI3 repressor. Here we identify a novel function for GLI3 repressor in controlling nephron number. During renal morphogenesis, HH signaling activity, assayed by expression of Ptc1-lacZ, is localized to ureteric cells of the medulla, but is undetectable in the cortex. Targeted inactivation of Smo, the HH effector, in the ureteric cell lineage causes no detectable abnormality in renal morphogenesis. The functional significance of absent HH signaling activity in cortical ureteric cells was determined by targeted deletion of Ptc1, the SMO inhibitor, in the ureteric cell lineage. Ptc1(-/-UB) mice demonstrate ectopic Ptc1-lacZ expression in ureteric branch tips and renal hypoplasia characterized by reduced kidney size and a paucity of mature and intermediate nephrogenic structures. Ureteric tip cells are remarkable for abnormal morphology and impaired expression of Ret and Wnt11, markers of tip cell differentiation. A finding of renal hypoplasia in Gli3(-/-) mice suggests a pathogenic role for reduced GLI3 repressor in the Ptc1(-/-UB) mice. Indeed, constitutive expression of GLI3 repressor via the Gli3(Delta699) allele in Ptc1(-/-UB) mice restores the normal pattern of HH signaling, and expression of Ret and Wnt11 and rescued the renal phenotype. Thus, GLI3 repressor controls nephron number by regulating ureteric tip cell expression of Wnt11 and Ret.


Subject(s)
Gene Expression Regulation, Developmental , Kidney/embryology , Kidney/metabolism , Kruppel-Like Transcription Factors/metabolism , Nephrons/embryology , Nephrons/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Ureter/metabolism , Wnt Proteins/metabolism , Animals , Apoptosis , Cell Lineage , Female , Male , Mice , Mice, Transgenic , Phenotype , Zinc Finger Protein Gli3
8.
Cancer Res ; 69(11): 4682-90, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487286

ABSTRACT

Subpopulations of tumorigenic cells have been identified in many human tumors, although these cells may not be very rare in some types of cancer. Here, we report that medulloblastomas arising from Patched-1-deficient mice contain a subpopulation of cells that show a neural precursor phenotype, clonogenic and multilineage differentiation capacity, activated Hedgehog signaling, wild-type Patched-1 expression, and the ability to initiate tumors following allogeneic orthotopic transplantation. The normal neural stem cell surface antigen CD15 enriches for the in vitro proliferative and in vivo tumorigenic potential from uncultured medulloblastomas, supporting the existence of a cancer stem cell hierarchy in this clinically relevant mouse model of cancer.


Subject(s)
Cerebellar Neoplasms/pathology , Lewis X Antigen/metabolism , Medulloblastoma/pathology , Multipotent Stem Cells/pathology , Neoplastic Stem Cells/pathology , Receptors, Cell Surface/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Embryo, Mammalian , Fucosyltransferases/metabolism , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Models, Biological , Multipotent Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Patched Receptors , Patched-1 Receptor , Phenotype , Receptors, Cell Surface/metabolism
9.
Dev Biol ; 308(2): 547-60, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17631878

ABSTRACT

Hedgehog (Hh) signaling is conserved from flies to humans and is indispensable in embryogenesis and adulthood. Patched (Ptc) encodes a receptor for Hh ligands and functions as a tumor suppressor. PTCH1 mutations in humans are found in basal cell carcinoma (BCC) and irradiated Ptc1(+/-) mice recapitulate this phenotype. However, due to embryonic lethality associated with the Ptc1 null mutation, its normal function in embryonic and adult skin remains unknown. Here we describe the epidermal phenotypes of a spontaneous and viable allele of Ptc1, Ptc1(mes), in which the C-terminal domain (CTD) is truncated. Ptc1(mes/mes) embryos display normal epidermal and hair follicle development. Postnatal Ptc1(mes/mes) skin displays severe basal cell layer hyperplasia and increased proliferation, while stratification of the suprabasal layers is mostly normal. Interestingly, truncation of the Ptc1 CTD did not result in skin tumors. However, long term labeling studies revealed a greater than three-fold increase in label-retaining cells in the interfollicular epidermis of Ptc1(mes/mes) adults, indicating possible expansion of the epidermal stem cell compartment. Increased expression of regulators of epidermal homeostasis, c-Myc and p63, was also observed in Ptc1(mes/mes) adult skin. These results suggest that the CTD of Ptc1 is involved in regulating epidermal homeostasis in mature skin.


Subject(s)
Receptors, Cell Surface/genetics , Skin Abnormalities/genetics , Skin Abnormalities/pathology , Stem Cells/pathology , Animals , Base Sequence , Cell Proliferation , DNA Primers/genetics , Gene Expression Regulation, Developmental , Genes, myc , Hedgehog Proteins/metabolism , Homeostasis , Humans , Hyperplasia , In Situ Hybridization , Mice , Mice, Mutant Strains , Patched Receptors , Patched-1 Receptor , Phenotype , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/physiology , Sequence Deletion , Signal Transduction , Skin/cytology , Skin/embryology , Skin/growth & development , Skin/metabolism , Skin Abnormalities/metabolism , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL