Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Signal ; 16(807): eadd6834, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37847757

RESUMO

Hedgehog (Hh) signaling controls growth and patterning during embryonic development and homeostasis in adult tissues. Hh binding to the receptor Patched (Ptc) elicits intracellular signaling by relieving Ptc-mediated inhibition of the transmembrane protein Smoothened (Smo). We uncovered a role for the lipid phosphatidic acid (PA) in the regulation of the Hh pathway in Drosophila melanogaster. Deleting the Ptc C-terminal tail or mutating the predicted PA-binding sites within it prevented Ptc from inhibiting Smo in wing discs and in cultured cells. The C-terminal tail of Ptc directly interacted with PA in vitro, an association that was reduced by Hh, and increased the amount of PA at the plasma membrane in cultured cells. Smo also interacted with PA in vitro through a binding pocket located in the transmembrane region, and mutating residues in this pocket reduced Smo activity in vivo and in cells. By genetically manipulating PA amounts in vivo or treating cultured cells with PA, we demonstrated that PA promoted Smo activation. Our findings suggest that Ptc may sequester PA in the absence of Hh and release it in the presence of Hh, thereby increasing the amount of PA that is locally available to promote Smo activation.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened/genética , Receptor Smoothened/metabolismo
2.
Int J Mol Sci ; 24(10)2023 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-37240278

RESUMO

Clear cell renal cell carcinoma (ccRCC) is the deadliest neoplasm of the urinary tract, and we are still far from completely understanding ccRCC development and treatment. The renal tissue paraffin blocks (20) of patients with ccRCC were collected at the University Hospital in Split from 2019 to 2020, and tissue sections were stained with patched (PTCH), anti-smoothened (SMO) and anti-Sonic Hedgehog (SHH) antibodies. SHH was highly expressed (31.9%) in grade 1 tumour, it being higher than all other grades and the control (p < 0.001-p < 0.0001). The trend of a linear decrease in the expression of SHH was observed with the progression of the tumour grade (p < 0.0001). PTCH expression was significantly lower in grades 1 and 2 in comparison to the control (p < 0.01) and grade 4 (p < 0.0001). A significant increase in the expression of SMO was found in grade 4 compared to all other grades (p < 0.0001) and the control (p < 0.001). The strong expression of SHH was observed in carcinoma cells of the G1 stage with a diffuse staining pattern (>50% of neoplastic cells). Stroma and/or inflammatory infiltrate display no staining and no expression of SHH in G1 and G2, while mild focal staining (10-50% of neoplastic cells) was observed in G3 and G4. Patients with high PTCH and low SMO expression had significant time survival differences (p = 0.0005 and p = 0.029, respectively). Therefore, high levels of PTCH and low levels of SMO expression are important markers of better survival rates in ccRCC patients.


Assuntos
Carcinoma de Células Renais , Carcinoma , Neoplasias Renais , Humanos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Carcinoma de Células Renais/genética , Receptores Patched/metabolismo , Transdução de Sinais , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Neoplasias Renais/genética , Receptor Smoothened/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
3.
Int J Mol Sci ; 24(3)2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36769003

RESUMO

Deleterious mutations in the X-linked Patched domain-containing 1 (PTCHD1) gene may account for up to 1% of autism cases. Despite this, the PTCHD1 protein remains poorly understood. Structural similarities to Patched family proteins point to a role in sterol transport, but this hypothesis has not been verified experimentally. Additionally, PTCHD1 has been suggested to be involved in Hedgehog signalling, but thus far, the experimental results have been conflicting. To enable a variety of biochemical and structural experiments, we developed a method for expressing PTCHD1 in Spodoptera frugiperda cells, solubilising it in glycol-diosgenin, and purifying it to homogeneity. In vitro and in silico experiments show that PTCHD1 function is not interchangeable with Patched 1 (PTCH1) in canonical Hedgehog signalling, since it does not repress Smoothened in Ptch1-/- mouse embryonic fibroblasts and does not bind Sonic Hedgehog. However, we found that PTCHD1 binds cholesterol similarly to PTCH1. Furthermore, we identified 13 PTCHD1-specific protein interactors through co-immunoprecipitation and demonstrated a link to cell stress responses and RNA stress granule formation. Thus, our results support the notion that despite structural similarities to other Patched family proteins, PTCHD1 may have a distinct cellular function.


Assuntos
Fibroblastos , Proteínas Hedgehog , Animais , Camundongos , Proteínas Hedgehog/metabolismo , Fibroblastos/metabolismo , Receptores Patched/metabolismo , Transdução de Sinais , Colesterol/metabolismo , Proteínas de Membrana/metabolismo
4.
Acta Histochem ; 124(1): 151835, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34979374

RESUMO

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.


Assuntos
Proteínas Hedgehog , Desenvolvimento Maxilofacial , Receptor Patched-2 , Animais , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Camundongos , Receptores Patched/metabolismo , Receptor Patched-2/genética , Receptor Patched-2/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais
5.
Mol Metab ; 47: 101172, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33513436

RESUMO

OBJECTIVE: Astrocytes are glial cells proposed as the main Sonic hedgehog (Shh)-responsive cells in the adult brain. Their roles in mediating Shh functions are still poorly understood. In the hypothalamus, astrocytes support neuronal circuits implicated in the regulation of energy metabolism. In this study, we investigated the impact of genetic activation of Shh signaling on hypothalamic astrocytes and characterized its effects on energy metabolism. METHODS: We analyzed the distribution of gene transcripts of the Shh pathway (Ptc, Gli1, Gli2, and Gli3) in astrocytes using single molecule fluorescence in situ hybridization combined with immunohistofluorescence of Shh peptides by Western blotting in the adult mouse hypothalamus. Based on the metabolic phenotype, we characterized Glast-CreERT2-YFP-Ptc-/- (YFP-Ptc-/-) mice and their controls over time and under a high-fat diet (HFD) to investigate the potential effects of conditional astrocytic deletion of the Shh receptor Patched (Ptc) on metabolic efficiency, insulin sensitivity, and systemic glucose metabolism. Molecular and biochemical assays were used to analyze the alteration of key pathways modulating energy metabolism, insulin sensitivity, glucose uptake, and inflammation. Primary astrocyte cultures were used to evaluate a potential role of Shh signaling in astrocytic glucose uptake. RESULTS: Shh peptides were the highest in the hypothalamic extracts of adult mice and a large population of hypothalamic astrocytes expressed Ptc and Gli1-3 mRNAs. Characterization of Shh signaling after conditional Ptc deletion in the YFP-Ptc-/- mice revealed heterogeneity in hypothalamic astrocyte populations. Interestingly, activation of Shh signaling in Glast+ astrocytes enhanced insulin responsiveness as evidenced by glucose and insulin tolerance tests. This effect was maintained over time and associated with lower blood insulin levels and also observed under a HFD. The YFP-Ptc-/- mice exhibited a lean phenotype with the absence of body weight gain and a marked reduction of white and brown adipose tissues accompanied by increased whole-body fatty acid oxidation. In contrast, food intake, locomotor activity, and body temperature were not altered. At the cellular level, Ptc deletion did not affect glucose uptake in primary astrocyte cultures. In the hypothalamus, activation of the astrocytic Shh pathway was associated with the upregulation of transcripts coding for the insulin receptor and liver kinase B1 (LKB1) after 4 weeks and the glucose transporter GLUT-4 after 32 weeks. CONCLUSIONS: Here, we define hypothalamic Shh action on astrocytes as a novel master regulator of energy metabolism. In the hypothalamus, astrocytic Shh signaling could be critically involved in preventing both aging- and obesity-related metabolic disorders.


Assuntos
Astrócitos/metabolismo , Glucose/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Envelhecimento , Animais , Astrócitos/patologia , Metabolismo Energético/genética , Células HEK293 , Proteínas Hedgehog/genética , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Neurônios/metabolismo , Obesidade , Receptores Patched/deficiência , Receptores Patched/genética , Transdução de Sinais , Ativação Transcricional
6.
Genome Biol ; 22(1): 33, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33446260

RESUMO

BACKGROUND: Aberrant activation of the Hedgehog pathway drives tumorigenesis of many cancers, including glioblastoma. However, the sensitization mechanism of the G protein-coupled-like receptor smoothened (SMO), a key component of Hedgehog signaling, remains largely unknown. RESULTS: In this study, we describe a novel protein SMO-193a.a. that is essential for Hedgehog signaling activation in glioblastoma. Encoded by circular SMO (circ-SMO), SMO-193a.a. is required for sonic hedgehog (Shh) induced SMO activation, via interacting with SMO, enhancing SMO cholesterol modification, and releasing SMO from the inhibition of patched transmembrane receptors. Deprivation of SMO-193a.a. in brain cancer stem cells attenuates Hedgehog signaling intensity and suppresses self-renewal, proliferation in vitro, and tumorigenicity in vivo. Moreover, circ-SMO/SMO-193a.a. is positively regulated by FUS, a direct transcriptional target of Gli1. Shh/Gli1/FUS/SMO-193a.a. form a positive feedback loop to sustain Hedgehog signaling activation in glioblastoma. Clinically, SMO-193a.a. is more specifically expressed in glioblastoma than SMO and is relevant to Gli1 expression. Higher expression of SMO-193a.a. predicts worse overall survival of glioblastoma patients, indicating its prognostic value. CONCLUSIONS: Our study reveals that SMO-193a.a., a novel protein encoded by circular SMO, is critical for Hedgehog signaling, drives glioblastoma tumorigenesis and is a novel target for glioblastoma treatment.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Proteínas Hedgehog/genética , RNA Circular/genética , Transdução de Sinais/genética , Receptor Smoothened/genética , Animais , Neoplasias Encefálicas/patologia , Proliferação de Células , Transformação Celular Neoplásica , Modelos Animais de Doenças , Feminino , Glioblastoma/patologia , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptores Patched/metabolismo , Receptor Smoothened/metabolismo , Células-Tronco
7.
Proc Natl Acad Sci U S A ; 117(46): 28838-28846, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33139559

RESUMO

Activation of the Hedgehog pathway may have therapeutic value for improved bone healing, taste receptor cell regeneration, and alleviation of colitis or other conditions. Systemic pathway activation, however, may be detrimental, and agents amenable to tissue targeting for therapeutic application have been lacking. We have developed an agonist, a conformation-specific nanobody against the Hedgehog receptor Patched1 (PTCH1). This nanobody potently activates the Hedgehog pathway in vitro and in vivo by stabilizing an alternative conformation of a Patched1 "switch helix," as revealed by our cryogenic electron microscopy structure. Nanobody-binding likely traps Patched in one stage of its transport cycle, thus preventing substrate movement through the Patched1 sterol conduit. Unlike the native Hedgehog ligand, this nanobody does not require lipid modifications for its activity, facilitating mechanistic studies of Hedgehog pathway activation and the engineering of pathway activating agents for therapeutic use. Our conformation-selective nanobody approach may be generally applicable to the study of other PTCH1 homologs.


Assuntos
Receptor Patched-1/agonistas , Receptor Patched-1/metabolismo , Receptor Patched-1/ultraestrutura , Animais , Microscopia Crioeletrônica/métodos , Proteínas Hedgehog/metabolismo , Humanos , Receptores Patched/metabolismo , Transdução de Sinais/fisiologia , Anticorpos de Domínio Único/farmacologia
8.
Int J Mol Sci ; 21(20)2020 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-33066274

RESUMO

Gorlin syndrome is a skeletal disorder caused by a gain of function mutation in Hedgehog (Hh) signaling. The Hh family comprises of many signaling mediators, which, through complex mechanisms, play several important roles in various stages of development. The Hh information pathway is essential for bone tissue development. It is also the major driver gene in the development of basal cell carcinoma and medulloblastoma. In this review, we first present the recent advances in Gorlin syndrome research, in particular, the signaling mediators of the Hh pathway and their functions at the genetic level. Then, we discuss the phenotypes of mutant mice and Hh signaling-related molecules in humans revealed by studies using induced pluripotent stem cells.


Assuntos
Síndrome do Nevo Basocelular/genética , Testes Genéticos/métodos , Animais , Síndrome do Nevo Basocelular/diagnóstico , Síndrome do Nevo Basocelular/metabolismo , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Instabilidade Genômica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Receptores Patched/genética , Receptores Patched/metabolismo
9.
Dev Cell ; 54(1): 92-105.e5, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32544391

RESUMO

Evolutionarily conserved signaling pathways are crucial for adjusting growth, reproduction, and cell maintenance in response to altered environmental conditions or energy balance. However, we have an incomplete understanding of the signaling networks and mechanistic changes that coordinate physiological changes across tissues. We found that loss of the cAMP response element-binding protein (CREB) transcription factor significantly slows Caenorhabditis elegans' reproductive decline, an early hallmark of aging in many animals. Our results indicate that CREB acts downstream of the transforming growth factor ß (TGF-ß) Sma/Mab pathway in the hypodermis to control reproductive aging, and that it does so by regulating a Hedgehog-related signaling factor, WRT-10. Overexpression of hypodermal wrt-10 is sufficient to delay reproductive decline and oocyte quality deterioration, potentially acting via Patched-related receptors in the germline. This TGF-ß-CREB-Hedgehog signaling axis allows a key metabolic tissue to communicate with the reproductive system to regulate oocyte quality and the rate of reproductive decline.


Assuntos
Envelhecimento/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Reprodução , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteínas Hedgehog/genética , Oócitos/metabolismo , Receptores Patched/genética , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
10.
Cancer Res ; 80(10): 1970-1980, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32060146

RESUMO

The Sonic Hedgehog (SHH) pathway plays a key role in cancer. Alterations of SHH canonical signaling, causally linked to tumor progression, have become rational targets for cancer therapy. However, Smoothened (SMO) inhibitors have failed to show clinical benefit in patients with cancers displaying SHH autocrine/paracrine expression. We reported earlier that the SHH receptor Patched (PTCH) is a dependence receptor that triggers apoptosis in the absence of SHH through a pathway that differs from the canonical one, thus generating a state of dependence on SHH for survival. Here, we propose a dual function for SHH: its binding to PTCH not only activates the SHH canonical pathway but also blocks PTCH-induced apoptosis. Eighty percent, 64%, and 8% of human colon, pancreatic, and lung cancer cells, respectively, overexpressed SHH at transcriptional and protein levels. In addition, SHH-overexpressing cells expressed all the effectors of the PTCH-induced apoptotic pathway. Although the canonical pathway remained unchanged, autocrine SHH interference in colon, pancreatic, and lung cell lines triggered cell death through PTCH proapoptotic signaling. In vivo, SHH interference in colon cancer cell lines decreased primary tumor growth and metastasis. Therefore, the antitumor effect associated to SHH deprivation, usually thought to be a consequence of the inactivation of the canonical SHH pathway, is, at least in part, because of the engagement of PTCH proapoptotic activity. Together, these data strongly suggest that therapeutic strategies based on the disruption of SHH/PTCH interaction in SHH-overexpressing cancers should be explored. SIGNIFICANCE: Sonic Hedgehog-overexpressing tumors express PTCH-induced cell death effectors, suggesting that this death signaling could be activated as an antitumor strategy.


Assuntos
Apoptose/fisiologia , Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Receptores Patched/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Embrião de Galinha , Xenoenxertos , Humanos , Camundongos , Transdução de Sinais/fisiologia , Peixe-Zebra
11.
PLoS One ; 15(2): e0229362, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32078657

RESUMO

In the mature rodent brain, Sonic Hedgehog (Shh) signaling regulates stem and progenitor cell maintenance, neuronal and glial circuitry and brain repair. However, the sources and distribution of Shh mediating these effects are still poorly characterized. Here, we report in the adult mouse brain, a broad expression pattern of Shh recognized by the specific monoclonal C9C5 antibody in a subset (11-12%) of CC1+ mature oligodendrocytes that do not express carbonic anhydrase II. These cells express also Olig2 and Sox10, two oligodendrocyte lineage-specific markers, but not PDGFRα, a marker of oligodendrocyte progenitors. In agreement with oligodendroglial cells being a source of Shh in the adult mouse brain, we identify Shh transcripts by single molecule fluorescent in situ hybridization in a subset of cells expressing Olig2 and Sox10 mRNAs. These findings also reveal that Shh expression is more extensive than originally reported. The Shh-C9C5-associated signal labels the oligodendroglial cell body and decorates by intense puncta the processes. C9C5+ cells are distributed in a grid-like manner. They constitute small units that could deliver locally Shh to its receptor Patched expressed in GFAP+ and S100ß+ astrocytes, and in HuC/D+ neurons as shown in PtcLacZ/+ reporter mice. Postnatally, C9C5 immunoreactivity overlaps the myelination peak that occurs between P10 and P20 and is down regulated during ageing. Thus, our data suggest that C9C5+CC1+ oligodendroglial cells are a source of Shh in the mouse postnatal brain.


Assuntos
Anticorpos Monoclonais/imunologia , Encéfalo/metabolismo , Proteínas Hedgehog/imunologia , Proteínas Hedgehog/metabolismo , Neurônios/metabolismo , Oligodendroglia/metabolismo , Animais , Encéfalo/imunologia , Células Cultivadas , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/imunologia , Oligodendroglia/imunologia , Receptores Patched/imunologia , Receptores Patched/metabolismo
12.
Cell Death Differ ; 27(4): 1286-1299, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31527798

RESUMO

Sonic hedgehog (SHH) signaling is crucial for the maintenance of the physiological self-renewal of granule neuron progenitor cells (GNPs) during cerebellar development, and its dysregulation leads to oncogenesis. However, how SHH signaling is controlled during cerebellar development is poorly understood. Here, we show that Trim32, a cell fate determinant, is distributed asymmetrically in the cytoplasm of mitotic GNPs, and that genetic knockout of Trim32 keeps GNPs at a proliferating and undifferentiated state. In addition, Trim32 knockout enhances the incidence of medulloblastoma (MB) formation in the Ptch1 mutant mice. Mechanistically, Trim32 binds to Gli1, an effector of SHH signaling, via its NHL domain and degrades the latter through its RING domain to antagonize the SHH pathway. These findings provide a novel mechanism that Trim32 may be a vital cell fate regulator by antagonizing the SHH signaling to promote GNPs differentiation and a tumor suppressor in MB formation.


Assuntos
Carcinogênese/metabolismo , Carcinogênese/patologia , Cerebelo/embriologia , Cerebelo/metabolismo , Proteínas Hedgehog/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo , Animais , Animais Recém-Nascidos , Proliferação de Células , Células HEK293 , Humanos , Meduloblastoma/patologia , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Receptores Patched/metabolismo , Ligação Proteica , Domínios Proteicos , Transdução de Sinais , Ubiquitina-Proteína Ligases/química , Ubiquitinação
13.
Curr Opin Cell Biol ; 61: 31-38, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31369952

RESUMO

The Hedgehog (HH) pathway plays a pivotal role in regulating a diverse array of events from embryonic tissue patterning to adult organ self-renewal. Aberrant activation of the pathway is linked to carcinogenesis. Key factors in the HH pathway include the signaling ligand HH, the receptor Patched (PTCH), and the G-protein-coupled receptor-like transducer Smoothened (SMO). A long-lasting question about this pathway is how PTCH prevents SMO from being activated. Recent high-resolution structural studies provide insight into the molecular basis of HH recognition by PTCH. Moreover, cholesterol stands out as the endogenous ligand of SMO and acts by binding and/or covalently linking to SMO. In this review, we discuss current advances in HH signaling, the interplay of PTCH, SMO and cholesterol, and propose putative models of SMO activation by cholesterol binding and/or modification.


Assuntos
Colesterol/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Transdução de Sinais , Receptor Smoothened/metabolismo , Animais , Humanos , Modelos Biológicos
14.
Curr Opin Struct Biol ; 57: 204-214, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31247512

RESUMO

The Hedgehog (HH) signalling pathway is a cell-cell communication system that controls the patterning of multiple tissues during embryogenesis in metazoans. In adults, HH signals regulate tissue stem cells and regenerative responses. Abnormal signalling can cause birth defects and cancer. The HH signal is received on target cells by Patched (PTCH1), the receptor for HH ligands, and then transmitted across the plasma membrane by Smoothened (SMO). Recent structural and biochemical studies have pointed to a sterol lipid, likely cholesterol itself, as the elusive second messenger that communicates the HH signal between PTCH1 and SMO, thus linking ligand reception to transmembrane signalling.


Assuntos
Colesterol/metabolismo , Proteínas Hedgehog/metabolismo , Receptores Patched/química , Receptores Patched/metabolismo , Transdução de Sinais , Receptor Smoothened/química , Receptor Smoothened/metabolismo , Animais , Humanos , Coluna Vertebral
16.
Basic Clin Pharmacol Toxicol ; 124(6): 660-669, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30548093

RESUMO

Tongxinluo capsule (TXL), a Chinese prescription, has been extensively used for treating ischaemic cerebrovascular diseases in China. Studies have demonstrated that TXL protects the blood-brain barrier (BBB) after cerebral ischaemia. However, the underlying protective mechanisms are not fully elucidated. Enlightened by the critical role of sonic hedgehog (Shh) pathway in promoting BBB integrity through up-regulating tight junction (TJ) proteins, we examined whether the Shh pathway could mediate TXL-induced up-regulation of TJ proteins and subsequent protection against BBB disruption after stroke. Ischaemic stroke was induced in adult male C57BL/6J mice by permanent middle cerebral artery occlusion (pMCAO). The mice were orally administered TXL (3.0 g/kg) at 1, 3 and 21 hours after stroke. Meanwhile, cyclopamine, a specific Shh pathway inhibitor, was intraperitoneally injected at 1 and 21 hours after stroke. The following parameters were measured at 6 and 24 hours after pMCAO: BBB permeability; TJ proteins including occludin, claudin-5 and zonula occludens-1 (ZO-1); and Shh signalling molecules such as Shh, Patched, Smoothened (Smo) and Gli-1. Our results showed that TXL protected against BBB disruption at 6 and 24 hours after pMCAO, and cyclopamine partly reversed the protective effect of TXL on BBB. Meanwhile, cyclopamine blocked the effect of TXL-up-regulated expression of occludin, claudin-5 and ZO-1. Moreover, TXL up-regulated the expression of Shh derived from astrocytes, Patched, Smo and Gli-1, and thus activated the Shh pathway. And cyclopamine inhibited TXL-induced activation of the Shh pathway. Thus, our study demonstrates that the Shh pathway mediates TXL-induced protection against BBB disruption after ischaemic stroke.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Proteínas Hedgehog/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Alcaloides de Veratrum/farmacologia , Animais , Isquemia Encefálica/patologia , Claudina-5/metabolismo , Células Endoteliais/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Camundongos Endogâmicos C57BL , Ocludina/metabolismo , Receptores Patched/metabolismo , Permeabilidade/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened/metabolismo , Acidente Vascular Cerebral/patologia , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
17.
Nat Cell Biol ; 19(5): 550-557, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28436966

RESUMO

Microorganisms and their hosts share the same environment, and microbial metabolic molecules (metabolites) exert crucial effects on host physiology. Environmental factors not only shape the composition of the host's resident microorganisms, but also modulate their metabolism. However, the exact molecular relationship among the environment, microbial metabolites and host metabolism remains largely unknown. Here, we discovered that environmental methionine tunes bacterial methyl metabolism to regulate host mitochondrial dynamics and lipid metabolism in Caenorhabditis elegans through an endocrine crosstalk involving NR5A nuclear receptor and Hedgehog signalling. We discovered that methionine deficiency in bacterial medium decreases the production of bacterial metabolites that are essential for phosphatidylcholine synthesis in C. elegans. Reductions of diundecanoyl and dilauroyl phosphatidylcholines attenuate NHR-25, a NR5A nuclear receptor, and release its transcriptional suppression of GRL-21, a Hedgehog-like protein. The induction of GRL-21 consequently inhibits the PTR-24 Patched receptor cell non-autonomously, resulting in mitochondrial fragmentation and lipid accumulation. Together, our work reveals an environment-microorganism-host metabolic axis regulating host mitochondrial dynamics and lipid metabolism, and discovers NR5A-Hedgehog intercellular signalling that controls these metabolic responses with critical consequences for host health and survival.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiologia , Proteínas de Ligação a DNA/metabolismo , Escherichia coli/metabolismo , Proteínas Hedgehog/metabolismo , Lipogênese , Receptores Patched/metabolismo , Fosfatidilcolinas/biossíntese , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Ligação a DNA/genética , Escherichia coli/genética , Genótipo , Proteínas Hedgehog/genética , Interações Hospedeiro-Patógeno , Metionina/deficiência , Metionina/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/microbiologia , Dinâmica Mitocondrial , Receptores Patched/genética , Fenótipo , Fatores de Tempo , Fatores de Transcrição/genética
18.
J Invest Dermatol ; 137(1): 179-186, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27498049

RESUMO

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.


Assuntos
Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Animais , Epiderme/embriologia , Epiderme/metabolismo , Proteínas Hedgehog/genética , Humanos , Hibridização in Situ Fluorescente , Camundongos , Camundongos Transgênicos , Modelos Animais , Receptores Patched/genética , Receptor Patched-2/genética , Receptor Patched-2/metabolismo , Sensibilidade e Especificidade
19.
Proc Natl Acad Sci U S A ; 113(40): E5866-E5875, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27647915

RESUMO

The Hedgehog cell-cell signaling pathway is crucial for animal development, and its misregulation is implicated in numerous birth defects and cancers. In unstimulated cells, pathway activity is inhibited by the tumor suppressor membrane protein, Patched. Hedgehog signaling is triggered by the secreted Hedgehog ligand, which binds and inhibits Patched, thus setting in motion the downstream events in signal transduction. Despite its critical importance, the mechanism by which Hedgehog antagonizes Patched has remained unknown. Here, we show that vertebrate Patched1 inhibition is caused by direct, palmitate-dependent interaction with the Sonic Hedgehog ligand. We find that a short palmitoylated N-terminal fragment of Sonic Hedgehog binds Patched1 and, strikingly, is sufficient to inhibit it and to activate signaling. The rest of Sonic Hedgehog confers high-affinity Patched1 binding and internalization through a distinct binding site, but, surprisingly, it is not absolutely required for signaling. The palmitate-dependent interaction with Patched1 is specifically impaired in a Sonic Hedgehog mutant causing human holoprosencephaly, the most frequent congenital brain malformation, explaining its drastically reduced potency. The palmitate-dependent interaction is also abolished in constitutively inhibited Patched1 point mutants causing the Gorlin cancer syndrome, suggesting that they might adopt a conformation distinct from the wild type. Our data demonstrate that Sonic Hedgehog signals via the palmitate-dependent arm of a two-pronged contact with Patched1. Furthermore, our results suggest that, during Hedgehog signaling, ligand binding inhibits Patched by trapping it in an inactive conformation, a mechanism that explains the dramatically reduced activity of oncogenic Patched1 mutants.


Assuntos
Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Transdução de Sinais , Células 3T3 , Animais , Endocitose/efeitos dos fármacos , Holoprosencefalia/metabolismo , Holoprosencefalia/patologia , Humanos , Lipoilação , Camundongos , Modelos Moleculares , Mutação/genética , Oncogenes , Ácido Palmítico/metabolismo , Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
20.
Cell Death Differ ; 23(12): 2042-2053, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27564589

RESUMO

Cerebellar granule neuronal progenitors (GNPs) are the precursors of cerebellar granule cells (CGCs) and are believed to be the cell of origin for medulloblastoma (MB), yet the molecular mechanisms governing GNP neurogenesis are poorly elucidated. Here, we demonstrate that storkhead box 1 (Stox1), a forkhead transcriptional factor, has a pivotal role in cerebellar granule neurogenesis and MB suppression. Expression of Stox1 is upregulated along with GNP differentiation and repressed by activation of sonic hedgehog (SHH) signaling. Stox1 exerts its neurogenic and oncosuppressing effect via direct transcriptional repression of Math1, a basic helix-loop-helix transcription activator essential for CGC genesis. This study illustrates a SHH-Stox1-Math1 regulatory axis in normal cerebellar development and MB formation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Transporte/metabolismo , Neoplasias Cerebelares/genética , Cerebelo/metabolismo , Grânulos Citoplasmáticos/metabolismo , Meduloblastoma/genética , Neurogênese , Transcrição Gênica , Animais , Sequência de Bases , Diferenciação Celular , Proliferação de Células , Neoplasias Cerebelares/patologia , Cerebelo/crescimento & desenvolvimento , Técnicas de Silenciamento de Genes , Proteínas Hedgehog , Meduloblastoma/patologia , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Receptores Patched/deficiência , Receptores Patched/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...