Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Biochem Biophys Res Commun ; 627: 76-83, 2022 10 30.
Article in English | MEDLINE | ID: mdl-36027694

ABSTRACT

Mutations in the human protein patched homolog (PTCH) gene have been demonstrated to be associated with cancer development in several types of malignancy. However, the underlying mechanism of PTCH-associated cancer development remains poorly understood, to the best of our knowledge. In the present study, the expression of PTCH2 in glioma tumor tissues from The Cancer Genome Atlas (TCGA) database and clinical patients with glioma were measured. Reduced expression levels of PTCH2 were observed in patients with glioma with poor prognose. In vitro, overexpression of PTCH2 significantly suppressed the proliferation and invasion of the glioma cell lines, LN229 and U87-MG. Mechanistically, PTCH2 upregulated the expression of tumor suppressor PTEN, thereby leading to the suppression of pro-survival AKT signals in glioma. Reduced expression of PTEN and enhanced expression of AKT promoted glioma development in vitro and in vivo. Blockade of PTCH2/AKT signals efficiently strengthened the anticancer effects of chemotherapy and prolonged the survival time in tumor-bearing mice, which provided a novel insight into potential treatment strategies for glioma in the clinic.


Subject(s)
Glioma , Proto-Oncogene Proteins c-akt , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioma/pathology , Humans , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics
2.
Acta Histochem ; 124(1): 151835, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34979374

ABSTRACT

Embryogenesis is modulated by numerous complex signaling cascades, which are essential for normal development. The Hedgehog (Hh) signaling pathway is part of these central cascades. As a homolog of Patched (Ptch)-1, Ptch2 initially did not appear to be as important as Ptch1. Recent reports have revealed that Ptch2 plays a crucial role in ligand-dependent feedback inhibition of Hh signaling in vertebrates. The role of Ptch2 in facial development remains unclear. Here, we investigated the detailed expression pattern of Ptch2 during craniofacial development in murine embryos based on in situ hybridization (ISH) studies of whole-mounts and sections, immunohistochemistry (IHC), and quantitative real-time PCR. We found that both Ptch2 mRNA and protein expression increased in a dynamic pattern in the facial development at mouse embryonic days 11-14.5. Moreover, distinct expression of Ptch2 was observed in the structures of the facial region, such as the tooth germ, Meckel's cartilage, and the follicles of vibrissae. These data, combined with our work in the macrostomia family, suggest that Ptch2 may play a critical role in facial development.


Subject(s)
Hedgehog Proteins , Maxillofacial Development , Patched-2 Receptor , Animals , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Patched Receptors/metabolism , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction
3.
Bioengineered ; 12(2): 12123-12134, 2021 12.
Article in English | MEDLINE | ID: mdl-34873972

ABSTRACT

The molecular mechanism that triggers polycystic ovary syndrome (PCOS) is mysterious. Abnormal development of ovarian granulosa cells (GCs) is one of the causes of PCOS. Herein, our study was carried out using RNA-seq to detect the different gene expression levels in ovarian GCs between three patients with PCOS and four normal controls. To verify the RNA-seq data, GCs from 22 patients with PCOS and 21 controls with normal ovulation were collected to perform the RT-PCR analysis. Hedgehog signaling pathway (Hh) members, Ihh and Ptch2 were abnormally highly expressed in the PCOS tissue (PT). The qPCR also indicated that the expression levels of Hh signaling pathway downstream members, Ptch1, Gli1, and Gli2 in the PT were significantly higher than those in the normal tissue (NT). Besides, the expression of TNF-α mRNA in PCOS patients was higher than that in the control group. Through the chromatin immunoprecipitation assay (ChIP), we found that the Gli1-IP-DNA enriched from the granular cells of PCOS patients was higher than that of the control group. Finally, the Hh signaling pathway inhibitor, cyclopamine, can decrease the apoptosis of PCOS ovarian granulosa cells. These results suggest that abnormal activation of Hh signaling pathway, especially Ihh signal, may have a profound influence on PCOS.


Subject(s)
Granulosa Cells/metabolism , Hedgehog Proteins/metabolism , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Signal Transduction , Adult , Apoptosis/genetics , Base Sequence , Case-Control Studies , Cells, Cultured , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation , Hedgehog Proteins/genetics , Humans , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , Polycystic Ovary Syndrome/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Treatment Outcome , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Young Adult
4.
Dev Biol ; 477: 177-190, 2021 09.
Article in English | MEDLINE | ID: mdl-34038742

ABSTRACT

Teleost fish fins, like all vertebrate limbs, comprise a series of bones laid out in characteristic pattern. Each fin's distal bony rays typically branch to elaborate skeletal networks providing form and function. Zebrafish caudal fin regeneration studies suggest basal epidermal-expressed Sonic hedgehog (Shh) promotes ray branching by partitioning pools of adjacent pre-osteoblasts. This Shh role is distinct from its well-studied Zone of Polarizing Activity role establishing paired limb positional information. Therefore, we investigated if and how Shh signaling similarly functions during developmental ray branching of both paired and unpaired fins while resolving cellular dynamics of branching by live imaging. We found shha is expressed uniquely by basal epidermal cells overlying pre-osteoblast pools at the distal aspect of outgrowing juvenile fins. Lateral splitting of each shha-expressing epidermal domain followed by the pre-osteoblast pools precedes overt ray branching. We use ptch2:Kaede fish and Kaede photoconversion to identify short stretches of shha+basal epidermis and juxtaposed pre-osteoblasts as the Shh/Smoothened (Smo) active zone. Basal epidermal distal collective movements continuously replenish each shha+domain with individual cells transiently expressing and responding to Shh. In contrast, pre-osteoblasts maintain Shh/Smo activity until differentiating. The Smo inhibitor BMS-833923 prevents branching in all fins, paired and unpaired, with surprisingly minimal effects on caudal fin initial skeletal patterning, ray outgrowth or bone differentiation. Staggered BMS-833923 addition indicates Shh/Smo signaling acts throughout the branching process. We use live cell tracking to find Shh/Smo restrains the distal movement of basal epidermal cells by apparent 'tethering' to pre-osteoblasts. We propose short-range Shh/Smo signaling promotes these heterotypic associations to couple instructive basal epidermal collective movements to pre-osteoblast repositioning as a unique mode of branching morphogenesis.


Subject(s)
Animal Fins/embryology , Epidermal Cells/physiology , Epidermis/embryology , Hedgehog Proteins/physiology , Morphogenesis , Zebrafish Proteins/physiology , Animal Fins/cytology , Animal Fins/metabolism , Animals , Benzamides/pharmacology , Cell Movement , Epidermis/metabolism , Patched-2 Receptor/metabolism , Quinazolines/pharmacology , Signal Transduction/drug effects , Smoothened Receptor/physiology , Zebrafish
5.
Clin Dermatol ; 38(4): 467-476, 2020.
Article in English | MEDLINE | ID: mdl-32972605

ABSTRACT

Basal cell nevus syndrome, also known as Gorlin syndrome, is a hereditary cancer syndrome associated with multiple basal cell carcinomas, congenital defects, and nondermatologic tumors. This disease is autosomal dominant with variable expressivity and is caused by abnormalities in the sonic hedgehog signaling pathway. Management requires a multidisciplinary approach and should include the biopsychosocial needs of patients and their families. Genetic testing is necessary to confirm an unclear diagnosis, evaluate at-risk relatives, and assist with family planning.


Subject(s)
Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/therapy , Molecular Targeted Therapy , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/therapy , Adult , Basal Cell Nevus Syndrome/diagnosis , Basal Cell Nevus Syndrome/pathology , Female , Genetic Testing , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Interdisciplinary Communication , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/pathology , Patched-1 Receptor/genetics , Patched-1 Receptor/metabolism , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , Patient Care Team , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Skin/pathology , Young Adult
6.
Lab Invest ; 100(9): 1208-1222, 2020 09.
Article in English | MEDLINE | ID: mdl-32457352

ABSTRACT

Aberrant activation of the hedgehog (Hh) signaling pathway is associated with the formation of medulloblastoma (MB), the most common malignant pediatric brain tumor. However, tumor cells from human and mouse MB can not be passaged or preserved after being adherently cultured. Moreover, Hh signaling in MB cells is inactivated in such culture. Here we demonstrate that MB cells are capable of forming tumoroids (tumor spheroids) in vitro under optimized conditions, which can be further passaged and cryopreserved. More importantly, MB cells maintain Hh pathway activation and cell proliferation in tumoroids. Our studies further reveal that tumoroids-forming capacity of MB cells relies on astrocytes, a major component of the MB microenvironment. Astrocytes facilitate the formation of MB tumoroids by secreting sonic hedgehog (Shh) and generating astrocyte-derived extracellular matrix. These findings demonstrate the critical role of stromal astrocytes in supporting the survival and proliferation of MB cells in vitro. This study establishes a valid model for long-term culture of primary MB cells, which could be greatly beneficial for future investigation of MB tumorigenicity and the development of improved approaches to treat MB.


Subject(s)
Astrocytes/metabolism , Cerebellar Neoplasms/genetics , Extracellular Matrix/metabolism , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Signal Transduction/genetics , Animals , Astrocytes/pathology , Cell Line, Tumor , Cerebellar Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice, Knockout , Mice, SCID , Mice, Transgenic , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , Tumor Microenvironment/genetics , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
7.
Nat Commun ; 11(1): 1994, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332736

ABSTRACT

Gas1 and Boc/Cdon act as co-receptors in the vertebrate Hedgehog signalling pathway, but the nature of their interaction with the primary Ptch1/2 receptors remains unclear. Here we demonstrate, using primordial germ cell migration in mouse as a developmental model, that specific hetero-complexes of Ptch2/Gas1 and Ptch1/Boc mediate the process of Smo de-repression with different kinetics, through distinct modes of Hedgehog ligand reception. Moreover, Ptch2-mediated Hedgehog signalling induces the phosphorylation of Creb and Src proteins in parallel to Gli induction, identifying a previously unknown Ptch2-specific signal pathway. We propose that although Ptch1 and Ptch2 functionally overlap in the sequestration of Smo, the spatiotemporal expression of Boc and Gas1 may determine the outcome of Hedgehog signalling through compartmentalisation and modulation of Smo-downstream signalling. Our study identifies the existence of a divergent Hedgehog signal pathway mediated by Ptch2 and provides a mechanism for differential interpretation of Hedgehog signalling in the germ cell niche.


Subject(s)
Chemotaxis/genetics , Gene Expression Regulation, Developmental , Germ Cells/physiology , Hedgehog Proteins/metabolism , Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , 3T3 Cells , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Embryo, Mammalian , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Knockout Techniques , Immunoglobulin G/metabolism , Intravital Microscopy , Male , Mice , Mice, Transgenic , Organ Culture Techniques , Patched-1 Receptor/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/genetics , Smoothened Receptor/metabolism , Time-Lapse Imaging , src-Family Kinases/metabolism
8.
PLoS One ; 14(11): e0225511, 2019.
Article in English | MEDLINE | ID: mdl-31756206

ABSTRACT

Basal cell carcinoma (BCC) is the most common human cancer, characterized by aberrant activation of the hedgehog (HH) signaling pathway resulting from mutations in the patched 1 (PTCH1) or smoothened (SMO) genes. In the present study, to uncover the expression profile of HH signaling-related molecules, we thoroughly examined the mRNA and protein expression levels of six molecules including GLI1, GLI2, PTCH1, PTCH2, SHH, and SMO in BCC and various other cutaneous tumors. Real-time PCR analysis demonstrated that BCC showed remarkably enhanced mRNA expression of all HH molecules, except SMO compared to other skin tumors. However, immunohistochemical analysis revealed that only GLI1 protein was specifically upregulated in BCC, while the other HH-related proteins did not show any significant differences between the tumors. Notably, other skin malignancies such as squamous cell carcinoma, sebaceous carcinoma, and malignant melanoma showed no GLI1 expression and there was no difference in GLI1 expression between the BCC subtypes. In addition, GLI1 and GLI2 expression were strongly associated with the hair follicle stem cell markers, LGR4 and LGR5, which are known target genes of the Wnt pathway. Our results suggest that GLI1 has the potential to be a diagnostically useful marker for differentiating BCC from other skin malignancies and an interaction between the HH and Wnt signaling pathways may be involved in the development of BCCs.


Subject(s)
Carcinoma, Basal Cell/pathology , Hedgehog Proteins/metabolism , Signal Transduction/genetics , Carcinoma, Basal Cell/metabolism , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Humans , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , RNA, Messenger/metabolism , Skin/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Stem Cells/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
9.
Cancer Lett ; 460: 65-74, 2019 Sep 28.
Article in English | MEDLINE | ID: mdl-31233836

ABSTRACT

Pds5b (precocious dissociation of sisters 5B) is involved in both tumorigenesis and cancer progression; however, the functions and molecular mechanisms of Pds5b in pancreatic cancer (PC) are unknown. Several approaches were conducted to investigate the molecular basis of Pds5b-related PC progression, including transfection, MTT, FACS, western blotting, wound healing assay, transwell chamber invasion assay, and immunohistochemical methods. Pds5b overexpression inhibited cell growth and induced apoptosis, whereas the inhibition of Pds5b promoted growth of PC cells. Moreover, Pds5b overexpression inhibited cell migration and invasion, while the downregulation of Pds5b enhanced cell motility. Furthermore, reduced Pds5b expression was associated with survival in PC patients. Mechanistically, Pds5b positively regulated the expression of Ptch2 to influence the Sonic hedgehog signaling pathway. Consistently, Ptch2 downregulation enhanced cell growth, migration, and invasion, while inhibiting cell apoptosis. Notably, the downregulation of Ptch2 abolished Pds5b-mediated anti-tumor activity in PC cells. Strikingly, Pds5b expression was positively associated with levels of Ptch2 in PC patient samples, suggesting that the Pds5b/Ptch2 axis regulates cell proliferation and invasion in PC cells. Our findings indicate that targeting Pds5b and Ptch2 may represent a novel therapeutic approach for PC.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/metabolism , Pancreatic Neoplasms/metabolism , Patched-2 Receptor/metabolism , Transcription Factors/metabolism , Apoptosis , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Patched-2 Receptor/genetics , Signal Transduction , Transcription Factors/genetics , Up-Regulation
10.
Stem Cells ; 37(9): 1238-1248, 2019 09.
Article in English | MEDLINE | ID: mdl-31145830

ABSTRACT

Continuous growth of the mouse incisor teeth is due to the life-long maintenance of epithelial stem cells (SCs) in their niche called cervical loop (CL). Several signaling factors regulate SC maintenance and/or their differentiation to achieve organ homeostasis. Previous studies indicated that Hedgehog signaling is crucial for both the maintenance of the SCs in the niche, as well as for their differentiation. How Hedgehog signaling regulates these two opposing cellular behaviors within the confinement of the CL remains elusive. In this study, we used in vitro organ and cell cultures to pharmacologically attenuate Hedgehog signaling. We analyzed expression of various genes expressed in the SC niche to determine the effect of altered Hedgehog signaling on the cellular hierarchy within the niche. These genes include markers of SCs (Sox2 and Lgr5) and transit-amplifying cells (P-cadherin, Sonic Hedgehog, and Yap). Our results show that Hedgehog signaling is a critical survival factor for SCs in the niche, and that the architecture and the diversity of the SC niche are regulated by multiple Hedgehog ligands. We demonstrated the presence of an additional Hedgehog ligand, nerve-derived Desert Hedgehog, secreted in the proximity of the CL. In addition, we provide evidence that Hedgehog receptors Ptch1 and Ptch2 elicit independent responses, which enable multimodal Hedgehog signaling to simultaneously regulate SC maintenance and differentiation. Our study indicates that the cellular hierarchy in the continuously growing incisor is a result of complex interplay of two Hedgehog ligands with functionally distinct Ptch receptors. Stem Cells 2019;37:1238-1248.


Subject(s)
Epithelial Cells/metabolism , Hedgehog Proteins/metabolism , Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , Stem Cell Niche , Stem Cells/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Epithelial Cells/cytology , Hedgehog Proteins/genetics , Incisor/cytology , Mice, Knockout , Mice, Transgenic , Models, Biological , Patched-1 Receptor/genetics , Patched-2 Receptor/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/genetics , Stem Cells/cytology
11.
Clin Oral Investig ; 23(1): 153-159, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29564556

ABSTRACT

AIMS: The aim of this study was to compare the clinical and demographic features of 62 patients presenting sporadic odontogenic keratocysts (OKCs) or OKCs associated with nevoid basal cell carcinoma syndrome (NBCCS). In conjunction with this, we also evaluated the immunohistochemical expression of Shh, Ptch1, Ptch2, Smo, Gli1, Gli2 and Gli3 proteins in 86 OKCs. By doing this, we add to the understanding of the biology of this type of lesion, providing tools that will help facilitate the early diagnosis of NBCCS in those patients where the first manifestation is that of OKCs. METHODS: This is a retrospective study; patients were classified into two groups: group 1 which consisted of those who were not affected by NBCCS (49 patients and 57 OKCs) and group 2 which consisted of those who were diagnosed with NBCCS (13 patients and 29 OKCs). The clinical and demographic features were studied and the immunohistochemical expression of Sonic Hedgehog proteins (Shh, Ptch1, Ptch2, Smo, Gli1, Gli2, and Gli3) was analyzed in all samples. RESULTS: There was an increase in the expression of three proteins in the syndromic OKC, when compared to that of sporadic cysts. Shh and Gli1 showed higher cytoplasmic expression, while Smo revealed stronger nuclear and cytoplasmic expressions. CONCLUSION AND CLINICAL RELEVANCE: Our findings suggest that the expression patterns of important Shh pathway proteins can represent valuable markers for early diagnosis of NBCCS-associated OKCs, as the major criterion for the diagnosis of NBCCS is currently based on the late appearance of basal cellular carcinomas. Thus, standardizing a new diagnostic tool for diagnosis of NBCCS could be of great importance in the identification of therapeutic targets. We therefore suggest, as based on our findings, that OKCs showing high expression of Shh, Smo, and Gli1 are potentially associated with NBCCS.


Subject(s)
Basal Cell Nevus Syndrome/metabolism , Hedgehog Proteins/metabolism , Jaw Neoplasms/metabolism , Odontogenic Cysts/metabolism , Signal Transduction/physiology , Adolescent , Adult , Aged , Biomarkers, Tumor/metabolism , Child , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , Retrospective Studies , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli3/metabolism
12.
J Biol Chem ; 293(43): 16583-16595, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30166346

ABSTRACT

Signaling through the Hedgehog (Hh) pathway is mediated by the Patched (Ptch) family of proteins. Although the vertebrate Ptch proteins Ptch1 and Ptch2 harbor two closely related transmembrane modules related to sterol-sensing domains (SSDs), the role of these closely related receptors in the Hh pathway are not equivalent. Ptch1 is essential for development and appears to be the principal receptor mediating responses to Hh ligands, whereas Ptch2 is nonessential, and its role in Hh-signaling remains ambiguous. We hypothesized that the SSDs of the Ptch proteins function as generic modules whose protein-specific activities are determined by the adjacent cytoplasmic and luminal domains. We first showed that individual N-terminal and C-terminal halves of Ptch1 associated noncovalently to mediate ligand-dependent regulation of Hh signaling. The analogous regions of Ptch2 also interacted noncovalently but did not repress the Hh pathway. However, the SSD of Ptch2 were capable of repressing Hh signaling, as determined using chimeric proteins where the SSDs of Ptch1 were replaced by those from Ptch2. Replacement of the SSDs of Ptch1 with the analogous regions from the cholesterol transporter NPC1 failed to produce a chimeric protein capable of Hh repression. Further refinement of the specific regions in Ptch1 and Ptch2 revealed that specific cytoplasmic domains of Ptch1 were necessary but not sufficient for repression of Hh signaling and that the two principal luminal domains of Ptch1 and Ptch2 were interchangeable. These data support a model where the SSDs of the Ptch family proteins exhibit generic activities and that the adjacent cytoplasmic and luminal domains determine their protein-specific activities.


Subject(s)
Cell Membrane/metabolism , Patched-1 Receptor/chemistry , Patched-1 Receptor/metabolism , Patched-2 Receptor/chemistry , Patched-2 Receptor/metabolism , Animals , Cell Membrane/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Knockout , Patched-1 Receptor/genetics , Patched-2 Receptor/genetics , Protein Binding , Protein Domains , Signal Transduction
13.
Mech Dev ; 153: 30-41, 2018 10.
Article in English | MEDLINE | ID: mdl-30144507

ABSTRACT

Sonic Hedgehog (Shh) signaling is characterized by non-cell autonomy; cells expressing Shh do not respond to the ligand. Here, we identify several Shh mutations that can activate the Hedgehog (Hh) pathway cell-autonomously. Cell-autonomous pathway activation requires the extracellular cysteine rich domain of Smoothened, but is otherwise independent of the Shh receptors Patched1 and -2. Many of the Shh mutants that gain activity fail to undergo auto processing resulting in the perdurance of the Shh pro-peptide, a form of Shh that is sufficient to activate the Hh response cell-autonomously. Our results demonstrate that Shh is capable of activating the Hh pathway via Smoothened, independently of Patched1/2, and that it harbors an intrinsic mechanism that prevents cell-autonomous activation of the Shh response.


Subject(s)
Gain of Function Mutation/genetics , Hedgehog Proteins/genetics , Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , Smoothened Receptor/metabolism , Amino Acid Sequence , Animals , Chickens , Holoprosencephaly/genetics , Holoprosencephaly/pathology , Luciferases/metabolism , Mice , Models, Biological , Neural Tube/embryology , Neural Tube/metabolism , Protein Domains , Smoothened Receptor/chemistry , Zinc Finger Protein GLI1/metabolism
14.
Cell Oncol (Dordr) ; 41(4): 427-437, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29869097

ABSTRACT

PURPOSE: Basal cell carcinoma (BCC) is one of the most common skin cancers, and is typically driven by an aberrantly activated Hedgehog (Hh) pathway. The Hh pathway is regulated by interactions between the Patched-1 (Ptch1) and Smoothened (Smo) receptors. Smo is an activating receptor and is subject to inhibition by Ptch1. Following ligand binding to Ptch1, its inhibitory action is relieved and pathway activation occurs. This receptor interaction is pivotal to restraining uncontrolled cellular growth. Both receptors have been found to be frequently mutated in BCCs. Ptch2 is a Ptch1 paralog that exhibits overlapping functions in both normal development and tissue homeostasis. As yet, its contribution to cancer growth is poorly defined. Here we set out to assess how Ptch2 inhibits BCC growth. METHODS: We used several in vitro readouts for transcriptional and chemotactic Hh signaling in BCC-derived ASZ001 cells, and a novel xenograft model to assess in vivo BCC tumor growth. Gene editing by TALEN was used to untangle the different Ptch2-dependent responses to its ligand sonic hedgehog (Shh). RESULTS: We first defined the signaling competence of Ptch2 in Ptch1-deficient ASZ001 cells in vitro, and found that Ptch2 ligand binding drives their migration rather than eliciting a transcriptional response. We found that subsequent targeting of Ptch2 abrogated the chemotaxic effect. Next, we tested the contribution of Ptch2 to in vivo tumor growth using a xenograft model and found that reduced Ptch function results in increased tumor growth, but that selective pressure appatently acts against complete Ptch2 ablation. CONCLUSIONS: We conclude that like Ptch1, Ptch2 exerts a tumor-suppressive function in BCC cells, and that after targeting of both paralogs, ligand-independent activation of the Hh pathway contributes to tumor growth.


Subject(s)
Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , Skin Neoplasms/metabolism , Animals , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Humans , Mice , Patched-1 Receptor/genetics , Patched-2 Receptor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Skin Neoplasms/genetics
15.
Eur Cytokine Netw ; 29(1): 27-35, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29748156

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by joints inflammation. One of the aggressive characteristics of RA fibroblast-like synoviocytes (FLS) is the tendency for migration in the local environment, which plays a central role in the RA pathogenesis. Tumor Necrosis Factor (TNF)-like cytokine 1A (TL1A) is a member of TNF superfamily, which has a role in autoimmunity and influences the RA-FLS behavior through TNF receptor 2 (TNFR2). We investigated the effect of TNF-like cytokine 1A (TL1A) on RA-FLS migration using patients' samples. Specifically, we examined the hedgehog signaling pathway which is a key regulator in chondrocyte growth and differentiation. We found that TL1A increased significantly the hedgehog homologue Indian hedgehog (IHH) and its receptor Patched 1, 2 (PTCH 1, 2) in RA-FLS. In addition, TL1A-stimulated RA-FLS promoted significantly IHH protein expression. However, both mRNA and protein levels decreased substantially after blocking TL1A with TNFR2 antagonist. The migratory property of RA-FLS was enhanced after stimulation of RA-FLS with TL1A, but was compromised following TL1A blockage. In conclusion, our study has revealed that TL1A modulated RA-FLS migration and Indian hedgehog signaling pathway using TNFR2.


Subject(s)
Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Movement , Fibroblasts/pathology , Hedgehog Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Synoviocytes/pathology , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Down-Regulation , Humans , Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors
16.
Cell Death Dis ; 9(5): 497, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29703892

ABSTRACT

Although many marker genes for germ cell differentiation have been identified, genes that specifically regulate primordial germ cell (PGC) generation are more difficult to determine. In the current study, we confirmed that C2EIP is a PGC marker gene that regulates differentiation by influencing the expression of pluripotency-associated genes such as Oct4 and Sox2. Knockout of C2EIP during embryonic development reduced PGC generation efficiency 1.5-fold, whereas C2EIP overexpression nearly doubled the generation efficiency both in vitro and in vivo. C2EIP encodes a cytoplasmic protein that interacted with PTCH2 at the intracellular membrane, promoted PTCH2 ubiquitination, activated the Hedgehog (HH) signaling pathway via competitive inhibition of the GPCR-like protein SMO, and positively regulated PGC generation. Activation and expression of C2EIP are regulated by the transcription factor STAT1, histone acetylation, and promoter methylation. Our data suggest that C2EIP is a novel, specific indicator of PGC generation whose gene product regulates embryonic stem cell differentiation by activating the HH signaling pathway via PTCH2 modification.


Subject(s)
Cell Differentiation , Embryonic Germ Cells/metabolism , Embryonic Stem Cells/metabolism , Hedgehog Proteins/metabolism , Patched-2 Receptor/metabolism , Acetylation , Animals , Chick Embryo , DNA Methylation , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Histones/metabolism , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Patched-2 Receptor/genetics , Phosphorylation , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Ubiquitination
17.
Clin Exp Dermatol ; 43(5): 518-524, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29424103

ABSTRACT

BACKGROUND: There is still much ambiguity in studies of Sonic hedgehog (Shh) pathways and its dysregulation. Some studies concerning the role of the Shh pathway in basal cell carcinoma (BCC) have been conducted, but there is a lack of studies about Shh pathway dysregulation under the influence of ultraviolet (UV)B radiation. AIM: To evaluate skin expression of Shh, Ptch1, Ptch2, Smo and Gli1 proteins in BCCs with and without the influence of UVB radiation. METHODS: In total, 34 healthy controls (HCs) and 42 patients with nodular BCC were recruited into the study. Patients were divided into five groups (A-E), depending on UVB dose received and BCC status. In all skin specimens, expression of Shh, Ptch1, Ptch2, Smo and Gli1 protein was evaluated. RESULTS: Comparing the BCC group with the HC group, there was significantly higher expression of Shh, Ptch1, Ptch2, Smo and Gli1 proteins. Expression of Ptch2, Smo and Gli1 was increased in response to UVB doses of 3 MED (minimal erythema dose), whereas expression of Ptch1 and Shh was unaffected. CONCLUSION: The lack of change in expression of Shh and Ptch1 after exposure to UVB suggests that the Shh pathway may be activated through a noncanonical pathway under the influence of strong UVB doses.


Subject(s)
Carcinoma, Basal Cell/metabolism , Hedgehog Proteins/metabolism , Skin Neoplasms/metabolism , Skin/metabolism , Ultraviolet Rays , Case-Control Studies , Humans , Middle Aged , Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , Radiation Dosage , Skin/radiation effects , Smoothened Receptor/metabolism , Zinc Finger Protein GLI1/metabolism
18.
EMBO Rep ; 18(11): 1922-1934, 2017 11.
Article in English | MEDLINE | ID: mdl-28887318

ABSTRACT

The Hh pathway controls many morphogenetic processes in metazoans and plays important roles in numerous pathologies and in cancer. Hh signaling is mediated by the activity of the Gli/Ci family of transcription factors. Several studies in Drosophila have shown that ubiquitination by the ubiquitin E3 ligases Slimb and Rdx(Hib) plays a crucial role in controlling Ci stability dependent on the levels of Hh signals. If Hh levels are low, Slimb adds K11- and K48-linked poly-ubiquitin chains on Ci resulting in partial degradation. Ubiquitin E2 enzymes are pivotal in determining the topologies of ubiquitin chains. However, which E2 enzymes participate in the selective ubiquitination-degradation of Ci remains elusive. Here, we find that the E2 enzyme UbcD1 negatively regulates Hh signaling activity in Drosophila wing disks. Genetic and biochemical analyses in wing disks and in cultured cells reveal that UbcD1 directly controls Ci stability. Interestingly, UbcD1 is found to be selectively involved in Slimb-mediated Ci degradation. Finally, we show that the homologs of UbcD1 play a conserved role in modulating Hh signaling in vertebrates.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Protein Processing, Post-Translational , Ubiquitin-Conjugating Enzymes/genetics , Zebrafish/genetics , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Conserved Sequence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Hedgehog Proteins/metabolism , Imaginal Discs/growth & development , Imaginal Discs/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , Polyubiquitin/genetics , Polyubiquitin/metabolism , Protein Stability , Proteolysis , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
19.
Dev Neurobiol ; 77(10): 1206-1220, 2017 10.
Article in English | MEDLINE | ID: mdl-28589698

ABSTRACT

Adult neurogenesis occurs more commonly in teleosts, represented by zebrafish, than in mammals. Zebrafish is therefore considered a suitable model to study adult neurogenesis, for which the regulatory molecular mechanisms remain little known. Our previous study revealed that neuroepithelial-like neural stem cells (NSCs) are located at the edge of the dorsomedial region. We also showed that Notch signaling inhibits NSC proliferation in this region. In the present study, we reported the expression of Wnt and Shh signaling components in this region of the optic tectum. Moreover, inhibitors of Wnt and Shh signaling suppressed NSC proliferation, suggesting that these pathways promote NSC proliferation. Shh is particularly required for maintaining Sox2-positive NSCs. Our experimental data also indicate the involvement of these signaling pathways in neural differentiation from NSCs. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1206-1220, 2017.


Subject(s)
Cell Proliferation/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Superior Colliculi/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Bromodeoxyuridine , Cell Proliferation/drug effects , Central Nervous System Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Imides/pharmacology , Immunohistochemistry , Membrane Proteins , Microscopy, Confocal , Microscopy, Fluorescence , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Patched-1 Receptor/metabolism , Patched-2 Receptor/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Quinolines/pharmacology , SOX Transcription Factors/metabolism , Superior Colliculi/drug effects , Thiadiazoles/pharmacology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , Zebrafish , Zebrafish Proteins/antagonists & inhibitors
20.
J Invest Dermatol ; 137(1): 179-186, 2017 01.
Article in English | MEDLINE | ID: mdl-27498049

ABSTRACT

By using the sensitivity of single-molecule fluorescent in situ hybridization, we have precisely quantified the levels and defined the temporal and spatial distribution of Hedgehog signaling activity during embryonic skin development and discovered that there is a Hedgehog signaling gradient along the proximal-distal axis of developing hair follicles. To explore the contribution of Hedgehog receptors Ptch1 and Ptch2 in establishing the epidermal signaling gradient, we quantitated the level of pathway activity generated in Ptch1- and Ptch1;Ptch2-deficient skin and defined the contribution of each receptor to regulation of the levels of Hedgehog signaling identified in wild-type skin. Moreover, we show that both the cellular phenotype and level of pathway activity featured in Ptch1;Ptch2-deficient cells faithfully recapitulates the Peak level of endogenous Hedgehog signaling detected at the base of developing follicles, where the concentration of endogenous Shh is predicted to be highest. Taken together, these data show that both Ptch1 and Ptch2 play a crucial role in sensing the concentration of Hedgehog ligand and regulating the appropriate dose-dependent response.


Subject(s)
Hedgehog Proteins/metabolism , Patched Receptors/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Epidermis/embryology , Epidermis/metabolism , Hedgehog Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Transgenic , Models, Animal , Patched Receptors/genetics , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL