Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260378

RESUMEN

Centrosome duplication and DNA replication are two pivotal events that higher eukaryotic cells use to initiate proliferation. While DNA replication is initiated through origin licensing, centrosome duplication starts with cartwheel assembly and is partly controlled by CP110. However, the upstream coordinator for both events has been, until now, a mystery. Here, we report that suppressor of fused protein (Sufu), a negative regulator of the Hedgehog (Hh) pathway playing a significant role in restricting the trafficking and function of glioma-related (Gli) proteins, acts as an upstream switch by facilitating CP110 phosphorylation by CDK2, promoting intranuclear Cdt1 degradation and excluding prereplication complex (pre-RC) components from chromosomes, independent of its canonical function in the Hh pathway. We found that Sufu localizes to both the centrosome and the nucleus and that knockout of Sufu induces abnormalities including centrosome amplification, increased nuclear size, multipolar spindle formation, and polyploidy. Serum stimulation promotes the elimination of Sufu from the centrosome by vesicle release at the ciliary tip and from the nucleus via protein degradation, which allows centrosome duplication and DNA replication to proceed. Collectively, this work reveals a mechanism through which Sufu negatively regulates the G1-S transition.


Asunto(s)
Centrosoma/metabolismo , Replicación del ADN , Proteínas Represoras/metabolismo , Animales , Proteínas de Unión a Calmodulina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Muerte Celular , Núcleo Celular/metabolismo , Cilios/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Vesículas Citoplasmáticas/metabolismo , Fibroblastos/metabolismo , Fase G1 , Células HEK293 , Células HeLa , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Mitosis , Mutación/genética , Fosforilación , Proteolisis , Proteínas Represoras/genética , Fase S
2.
Biochem Cell Biol ; 101(4): 284-293, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36821837

RESUMEN

Studies in the past decade have shown that lipid droplets stored in liver cells under starvation are encapsulated by autophagosomes and fused to lysosomes via the endocytic system. Autophagy responds to a variety of environmental factors inside and outside the cell, so it has a complex signal regulation network. To this end, we first explored the role of Hedgehog (Hh) in autophagy and lipid metabolism. Treatment of normal mouse liver cells with SAG and GDC-0449 revealed elevated phosphorylation of AMP-activated protein kinase (AMPK) and increased lipidation of LC3. SAG, and GDC-0449 were agonist and antagonist of Smoothened (Smo) in canonical Hh pathway, respectively, but they played a consistent role in the regulation of autophagy in hepatocytes. Moreover, SAG and GDC-0449 did not affect the expression of glioma-associated oncogene (Gli1) and patched 1, suggesting the absence of canonical Hh signaling in hepatocytes. We further knocked down the Smo and found that the effects of SAG and GDC-0449 disappeared, indicating that the non-canonical Smo pathway was involved in the regulation of autophagy in hepatocytes. In addition, SAG and GDC-0449 promoted lipid degradation and inhibited lipid production signals. Knockdown of Smo slowed down the rate of lipid degradation rather than Sufu or Gli1, indicating that Hh signaling regulated the lipid metabolism via Smo. In summary, activates AMPK via Smo to promote autophagy and lipid degradation.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Proteínas Hedgehog , Ratones , Animales , Proteínas Hedgehog/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Hepatocitos/metabolismo , Autofagia , Lípidos , Receptores Acoplados a Proteínas G/metabolismo
3.
J Biol Chem ; 292(4): 1351-1360, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-27979967

RESUMEN

The function of the primary cilia, which is assembled in most vertebrate cells, is achieved by transport in and out of kinds of signaling receptors. The BBSome protein complex could recognize and target membrane proteins to the cilia, but how the BBSome itself is transported into the cilia is poorly understood. Here we demonstrate that the centrosome protein Dzip1 mediates the assembly of the BBSome-Dzip1-PCM1 complex in the centriolar satellites (CS) at the G0 phase for ciliary translocation of the BBSome. Phosphorylation of Dzip1 at Ser-210 by Plk1 (polo-like kinase 1) during the G2 phase promotes disassembly of this complex, resulting in removal of Dzip1 and the BBSome from the CS. Inhibiting the kinase activity of Plk1 maintains the CS localization of the BBSome and Dzip1 at the G2 phase. Collectively, our findings reveal the cell cycle-dependent regulation of BBSome transport to the CS and highlight a potential mechanism that the BBSome-mediated signaling pathways are accordingly regulated during the cell cycle.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centriolos/metabolismo , Proteínas de Unión al ADN/metabolismo , Fase G2/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular/genética , Centriolos/genética , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas/genética , Quinasa Tipo Polo 1
4.
EMBO J ; 30(23): 4777-89, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045334

RESUMEN

TGF-ß signalling is regulated by post-translational modifications of Smad proteins to translate quantitative difference in ligand concentration into proportional transcriptional output. Previous studies in cell culture systems suggested that Smad ubiquitination regulatory factors (Smurfs) act in this regulation by targeting Smads for proteasomal degradation, but whether this mechanism operates under physiological conditions is not clear. Here, we generated mice harbouring a target-disrupted Smurf2 allele. Using primary mouse embryonic fibroblasts and dermal fibroblasts, we show that TGF-ß-mediated, Smad-dependent transcriptional responses are elevated in the absence of Smurf2. Instead of promoting poly-ubiquitination and degradation, we show that Smurf2 actually induces multiple mono-ubiquitination of Smad3 in vivo. Phosphorylation of T179, immediately upstream of the Smad3 PY motif, enhances Smurf2 and Smad3 interaction and Smad3 ubiquitination. We have mapped Smurf2-induced Smad3 ubiquitination sites to lysine residues at the MH2 domain, and demonstrate that Smad3 ubiquitination inhibits the formation of Smad3 complexes. Thus, our data support a model in which Smurf2 negatively regulates TGF-ß signalling by attenuating the activity of Smad3 rather than promoting its degradation.


Asunto(s)
Transducción de Señal/fisiología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina-Proteína Ligasas , Ubiquitinación/fisiología , Animales , Western Blotting , Fibroblastos/metabolismo , Ratones , Modelos Biológicos , Unión Proteica , Transcripción Genética/fisiología , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
JCI Insight ; 9(6)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38358805

RESUMEN

Suppressor of fused (SUFU) is widely regarded as a key negative regulator of the sonic hedgehog (SHH) morphogenic pathway and a known tumor suppressor of medulloblastoma (MB). However, we report here that SUFU expression was markedly increased in 75% of specimens compiled in a tissue array comprising 49 unstratified MBs. The SUFU and GLI1 expression levels in this MB array showed strong positive correlation, which was also identified in a large public data set containing 736 MBs. We further report that increasing Sufu gene dosage in mice caused preaxial polydactyly, which was associated with the expansion of the Gli3 domain in the anterior limb bud and heightened Shh signaling responses during embryonic development. Increasing Sufu gene dosage also led to accelerated cerebellar development and, when combined with ablation of the Shh receptor encoded by Patched1 (Ptch1), promoted MB tumorigenesis. These data reveal multifaceted roles of SUFU in promoting MB tumorigenesis by enhancing SHH signaling. This revelation clarifies potentially counterintuitive clinical observation of high SUFU expression in MBs and may pave way for novel strategies to reduce or reverse MB progression.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Polidactilia , Ratones , Animales , Meduloblastoma/genética , Meduloblastoma/patología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Transformación Celular Neoplásica/genética , Factores de Transcripción , Neoplasias Cerebelosas/genética , Polidactilia/genética
7.
Hum Mol Genet ; 20(19): 3725-37, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21653639

RESUMEN

The primary cilium is emerging as a crucial regulator of signaling pathways central to vertebrate development and human disease. We identified atrioventricular canal 1 (avc1), a mouse mutation that caused VACTERL association with hydrocephalus, or VACTERL-H. We showed that avc1 is a hypomorphic mutation of intraflagellar transport protein 172 (Ift172), required for ciliogenesis and Hedgehog (Hh) signaling. Phenotypically, avc1 caused VACTERL-H but not abnormalities in left-right (L-R) axis formation. Avc1 resulted in structural cilia defects, including truncated cilia in vivo and in vitro. We observed a dose-dependent requirement for Ift172 in ciliogenesis using an allelic series generated with Ift172(avc1) and Ift172(wim), an Ift172 null allele: cilia were present on 42% of avc1 mouse embryonic fibroblast (MEF) and 28% of avc1/wim MEFs, in contrast to >90% of wild-type MEFs. Furthermore, quantitative cilium length analysis identified two specific cilium populations in mutant MEFS: a normal population with normal IFT and a truncated population, 50% of normal length, with disrupted IFT. Cells from wild-type embryos had predominantly full-length cilia, avc1 embryos, with Hh signaling abnormalities but not L-R abnormalities, had cilia equally divided between full-length and truncated, and avc1/wim embryos, with both Hh signaling and L-R abnormalities, were primarily truncated. Truncated Ift172 mutant cilia showed defects of the distal ciliary axoneme, including disrupted IFT88 localization and Hh-dependent Gli2 localization. We propose a model in which mutation of Ift172 results in a specific class of abnormal cilia, causing disrupted Hh signaling while maintaining L-R axis determination, and resulting in the VACTERL-H phenotype.


Asunto(s)
Cardiopatías Congénitas/genética , Hidrocefalia/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Deformidades Congénitas de las Extremidades/genética , Ratones/genética , Proteínas Adaptadoras Transductoras de Señales , Alelos , Canal Anal/anomalías , Canal Anal/embriología , Canal Anal/metabolismo , Animales , Cilios/genética , Cilios/metabolismo , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Esófago/anomalías , Esófago/embriología , Esófago/metabolismo , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Hidrocefalia/embriología , Hidrocefalia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/anomalías , Riñón/embriología , Riñón/metabolismo , Deformidades Congénitas de las Extremidades/embriología , Deformidades Congénitas de las Extremidades/metabolismo , Ratones/metabolismo , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mutagénesis , Mutación , Transporte de Proteínas , Transducción de Señal/genética , Columna Vertebral/anomalías , Columna Vertebral/embriología , Columna Vertebral/metabolismo , Tráquea/anomalías , Tráquea/embriología , Tráquea/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
8.
Cell Biosci ; 13(1): 15, 2023 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-36683064

RESUMEN

BACKGROUND: Medulloblastoma (MB) is one of the most common malignant pediatric brain tumors. Metastasis and relapse are the leading causes of death in MB patients. The initiation of the SHH subgroup of MB (SHH-MB) is due to the aberrant activation of Sonic Hedgehog (Shh) signaling. However, the mechanisms for its metastasis are still unknown. RESULTS: AMP-dependent protein kinase (AMPK) restrains the activation of Shh signaling pathway, thereby impeding the proliferation of SHH-MB cells. More importantly, AMPK also hinders the growth and metastasis of SHH-MB cells by regulating NF-κB signaling pathway. Furthermore, Vismodegib and TPCA-1, which block the Shh and NF-κB pathways, respectively, synergistically restrained the growth, migration, and invasion of SHH-MB cells. CONCLUSIONS: This work demonstrates that AMPK functions through two signaling pathways, SHH-GLI1 and NF-κB. AMPK-NF-κB axis is a potential target for molecular therapy of SHH-MB, and the combinational blockade of NF-κB and Shh pathways confers synergy for SHH-MB therapy.

9.
J Biol Chem ; 286(15): 13502-11, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21317289

RESUMEN

Suppressor of fused (Sufu) is an essential negative regulator of the sonic hedgehog (Shh) pathway, but little is known about how Sufu itself is normally regulated. Here, we report that Sufu is phosphorylated at Ser-342 and Ser-346 by GSK3ß and cAMP-dependent protein kinase A (PKA), respectively, and phosphorylation at this dual site stabilizes Sufu against Shh signaling-induced degradation. We further show that localization of Sufu in the primary cilium is induced by Shh signaling and is required for the turnover of both phosphorylated and total Sufu. Perturbing Sufu phosphorylation with PKA inhibitors or replacing Ser-346 with alanine reduced the stay and replacing Ser-342 and Ser-346 with aspartic acid prolonged the stay of Sufu in the cilia. Finally, ciliary localization of Gli2/3 also required Smo and was similarly influenced by perturbations of PKA activity or mutations at the dual Sufu phosphorylation site. Thus, Shh likely induced trafficking of phospho-Sufu into the primary cilium in a complex with Gli2/3, and dephosphorylation triggered a retrograde export, allowing Sufu to be degraded by the ubiquitin-proteasome system.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Represoras/metabolismo , Sustitución de Aminoácidos , Animales , Cilios/genética , Cilios/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Mutación Missense , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación/fisiología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Transporte de Proteínas/fisiología , Proteínas Represoras/genética , Ubiquitina/genética , Ubiquitina/metabolismo , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
10.
J Biol Chem ; 285(34): 26599-607, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20534588

RESUMEN

Vertebrate muscle differentiation is coordinated by an intricate network of transcription factors requiring proliferating myogenic precursors to withdraw irreversibly from the cell cycle. Recent studies have implicated a large number of microRNAs exerting another layer of control in many aspects of muscle differentiation. By annealing to short recognition sequences in the 3'-untranslated region, microRNAs attenuate target gene expression through translation repression or mRNA degradation. Here, we show that miR-214 promotes myogenic differentiation in mouse C2C12 myoblasts at a step preceding the induction of p21 and myogenin. Blocking miR-214 function with a 2'-O-methylated double-stranded inhibitor maintained C2C12 cells in the active cell cycle, thereby inhibiting the myogenic differentiation. By global gene expression profiling, we identified the proto-oncogene N-ras as one of miR-214 targets. Furthermore, manipulating the N-Ras level with small interfering RNA or adenovirus-mediated forced expression either augmented or attenuated the effect of miR-214, respectively. Thus, our data uncovered a novel microRNA-mediated mechanism that controls myogenic differentiation.


Asunto(s)
Diferenciación Celular , Genes ras , MicroARNs/fisiología , Mitosis , Mioblastos/citología , Animales , Línea Celular , Regulación hacia Abajo/genética , Perfilación de la Expresión Génica , Ratones , Desarrollo de Músculos
11.
Cell Biosci ; 11(1): 46, 2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33653381

RESUMEN

BACKGROUND: Hedgehog signaling is essential to the regulation of embryonic development, tissue homeostasis, and stem cell self-renewal, making it a prime target for developing cancer therapeutics. Given the close link between aberrant Hedgehog signaling and cancers, many small molecular compounds have been developed to inhibit Smoothened, a key signal transducer of this pathway, for treating cancer and several such compounds have been approved by the United States Food and Drug Administration (GDC-0449 and LDE-225). However, acquired drug resistance has emerged as an important obstacle to the effective use of these first generation Hedgehog pathway blockers. Thus, new Smoothened inhibitors that can overcome such resistance is an urgent need going forward. RESULTS: We established the Smoothened/ßarrestin2-GFP high-throughput screening platform based on the mechanistic discovery of Hedgehog signaling pathway, and discovered several active small molecules targeting Smoothened including 0025A. Here we show that 0025A can block the translocation of ßarrestin2-GFP to Smoothened, displace Bodipy-cyclopamine binding to wild-type Smoothened or mutant Smoothened-D473H, reduce the accumulation of Smo on primary cilia and the expression of Gli upon Hedgehog stimulation. In addition, we show that 0025A can effectively suppress hair follicle morphogenesis and hair growth in mice. CONCLUSIONS: Our results demonstrate that 0025A is a potent antagonist targeting Smoothened wild-type and mutant receptors in the Hedgehog signaling pathway and may provide a new therapy for refractory cancers.

12.
Cell Death Discov ; 7(1): 120, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34021128

RESUMEN

Although E3 ligase Speckle type BTB/POZ protein (SPOP) promotes tumorigenesis by acting as a key regulatory hub in clear cell renal cell carcinoma (ccRCC), the detailed molecular mechanism remains unclear. Here, we demonstrate that a well-known tumor suppressor, Suppressor of Fused (SUFU), is downregulated by SPOP. Interestingly, this downregulation depends on cullin-3(Cul3)-SPOP E3 ligase, but SUFU is not a direct substrate of SPOP. Phosphatase and tensin homolog (PTEN), a ubiquitinated substrate of SPOP, is involved in SPOP-mediated SUFU reduction. Importantly, inhibition of SUFU leads to elevated SHH and WNT signaling, consequently rescuing the reduced proliferation, migration, and invasion abilities of ccRCC cells caused by SPOP-knockdown. Moreover, combinatorial treatment with SHH and WNT inhibitors shows more effective for suppressing ccRCC cell proliferation and aggressiveness. These findings demonstrate that a novel SPOP-PTEN-SUFU axis promotes ccRCC carcinogenesis by activating SHH and WNT pathway, providing a new treatment strategy for ccRCC.

13.
Biochim Biophys Acta Mol Cell Res ; 1868(12): 119124, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34419491

RESUMEN

Autophagy is a highly conservative self-digestion process to maintain intracellular homeostasis and to ensure the survival of cells under stress. Activation of Sonic Hedgehog (Shh) signaling depends on the normal endocytic degradation of pathway receptor Patched1 (Ptch1). It is unclear whether autophagy participates in the receptor endocytosis and modulates Shh signaling transduction. Here we found that blocking macroautophagy attenuates Shh signaling due to the failed transport of Smoothened (Smo) into primary cilia. At the upstream of Smo, Ptch1 was poly-ubiquitinated through K63-conjugated ubiquitin chains. Macroautophagy participates Shh-induced degradation of poly-ubiquitinated Ptch1, contributing to the activation of Shh signaling.


Asunto(s)
Autofagia , Proteínas Hedgehog/metabolismo , Receptor Patched-1/metabolismo , Proteolisis , Animales , Células Cultivadas , Cilios/metabolismo , Células HEK293 , Humanos , Ratones , Transducción de Señal , Receptor Smoothened/metabolismo , Ubiquitinación
14.
Cell Biosci ; 11(1): 47, 2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33653390

RESUMEN

BACKGROUND: Skeletal development and maintenance are complex processes known to be coordinated by multiple genetic and epigenetic signaling pathways. However, the role of long non-coding RNAs (lncRNAs), a class of crucial epigenetic regulatory molecules, has been under explored in skeletal biology. RESULTS: Here we report a young patient with short stature, hypothalamic dysfunction and mild macrocephaly, who carries a maternally inherited 690 kb deletion at Chr.1q24.2 encompassing a noncoding RNA gene, DNM3OS, embedded on the opposite strand in an intron of the DYNAMIN 3 (DNM3) gene. We show that lncRNA DNM3OS sustains the proliferation of chondrocytes independent of two co-cistronic microRNAs miR-199a and miR-214. We further show that nerve growth factor (NGF), a known factor of chondrocyte growth, is a key target of DNM3OS-mediated control of chondrocyte proliferation. CONCLUSIONS: This work demonstrates that DNM3OS is essential for preventing premature differentiation of chondrocytes required for bone growth through endochondral ossification.

15.
J Biomed Res ; 35(1): 21-35, 2020 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-33361643

RESUMEN

Colorectal cancer (CRC) is one of the most deadly cancers in the world with few reliable biomarkers that have been selected into clinical guidelines for prognosis of CRC patients. In this study, mRNA microarray datasets GSE113513, GSE21510, GSE44076, and GSE32323 were obtained from the Gene Expression Omnibus (GEO) and analyzed with bioinformatics to identify hub genes in CRC development. Differentially expressed genes (DEGs) were analyzed using the GEO2R tool. Gene ontology (GO) and KEGG analyses were performed through the DAVID database. STRING database and Cytoscape software were used to construct a protein-protein interaction (PPI) network and identify key modules and hub genes. Survival analyses of the DEGs were performed on GEPIA database. The Connectivity Map database was used to screen potential drugs. A total of 865 DEGs were identified, including 374 upregulated and 491 downregulated genes. These DEGs were mainly associated with metabolic pathways, pathways in cancer, cell cycle and so on. The PPI network was identified with 863 nodes and 5817 edges. Survival analysis revealed that HMMR, PAICS, ETFDH, and SCG2 were significantly associated with overall survival of CRC patients. And blebbistatin and sulconazole were identified as candidate drugs. In conclusion, our study found four hub genes involved in CRC, which may provide novel potential biomarkers for CRC prognosis, and two potential candidate drugs for CRC.

16.
Cell Death Dis ; 11(8): 686, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32826873

RESUMEN

Reversible phosphorylation of Suppressor of fused (Sufu) is essential for Sonic Hedgehog (Shh) signal transduction. Sufu is stabilized under dual phosphorylation of protein kinase A (PKA) and glycogen synthase kinase 3ß (GSK3ß). Its phosphorylation is reduced with the activation of Shh signaling. However, the phosphatase in this reversible phosphorylation has not been found. Taking advantage of a proteomic approach, we identified Protein phosphatase 4 regulatory subunit 2 (Ppp4r2), an interacting protein of Sufu. Shh signaling promotes the interaction of these two proteins in the nucleus, and Ppp4 also promotes dephosphorylation of Sufu, leading to its degradation and enhancing the Gli1 transcriptional activity. Finally, Ppp4-mediated dephosphorylation of Sufu promotes proliferation of medulloblastoma tumor cells, and expression of Ppp4 is positively correlated with up-regulation of Shh pathway target genes in the Shh-subtype medulloblastoma, underscoring the important role of this regulation in Shh signaling.


Asunto(s)
Proteínas Hedgehog/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fibroblastos/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas Hedgehog/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Meduloblastoma/genética , Ratones , Fosfoproteínas Fosfatasas/genética , Fosforilación , Proteómica/métodos , Proteínas Represoras/genética , Transducción de Señal/fisiología , Proteína con Dedos de Zinc GLI1/metabolismo
17.
Adv Cancer Res ; 101: 29-43, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19055941

RESUMEN

Originally identified as factors affecting Drosophila embryogenesis, the Hedgehog (Hh) pathway is one of the primary signaling systems that specify patterns of cell growth and differentiation during vertebrate development. Mutations in various components of this pathway frequently occur in tumors originated from the skin, cerebellum, and skeletal muscle, and abnormal pathway activity is associated with a subset of lung, digestive tract, pancreatic, and prostate cancers. Because of these potent biological activities, this pathway is negatively regulated at multiple levels to ensure appropriate signaling responses. Suppressor of fused (Sufu) is one such negative regulator of Hh signaling. Although not essential in Drosophila, Sufu is absolutely required for mouse embryonic development. Mutations of Sufu are associated with a childhood brain tumor in human and an increased susceptibility to the same type of cancer in the TP53 null background in mice, and RNAi-mediated silencing of Sufu is sufficient to activate the Hh signaling in cultured fibroblasts. All these data point to a central role of Sufu in controlling the vertebrate Hh signaling pathway; however, for years what exactly Sufu does in the Hh pathway and what controls its activity remains a deep mystery. This chapter will go over all studies curated in the PubMed database with Sufu as a main subject during the past 17 years, and attempt to provide a balanced view on Sufu gene and protein structure, activities in Drosophila as well as mammalian development, and its involvement in cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Proteínas Hedgehog/metabolismo , Proteínas Represoras/fisiología , Animales , Drosophila , Silenciador del Gen , Humanos , Ratones , Modelos Biológicos , Mutación , Neoplasias/metabolismo , Interferencia de ARN , Proteínas Represoras/química , Transducción de Señal , Especificidad de la Especie
18.
Dev Cell ; 48(2): 167-183.e5, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30554998

RESUMEN

SUFU alterations are common in human Sonic Hedgehog (SHH) subgroup medulloblastoma (MB). However, its tumorigenic mechanisms have remained elusive. Here, we report that loss of Sufu alone is unable to induce MB formation in mice, due to insufficient Gli2 activation. Simultaneous loss of Spop, an E3 ubiquitin ligase targeting Gli2, restores robust Gli2 activation and induces rapid MB formation in Sufu knockout background. We also demonstrated a tumor-promoting role of Sufu in Smo-activated MB (∼60% of human SHH MB) by maintaining robust Gli activity. Having established Gli2 activation as a key driver of SHH MB, we report a comprehensive analysis of its targetome. Furthermore, we identified Atoh1 as a target and molecular accomplice of Gli2 that activates core SHH MB signature genes in a synergistic manner. Overall, our work establishes the dual role of SUFU in SHH MB and provides mechanistic insights into transcriptional regulation underlying Gli2-mediated SHH MB tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Proteína Gli2 con Dedos de Zinc/genética , Animales , Proteínas Hedgehog/genética , Humanos , Meduloblastoma/genética , Ratones
20.
Mol Cell Biol ; 37(18)2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28630282

RESUMEN

The transcriptional output of the Sonic Hedgehog morphogenic pathway is orchestrated by three Krüppel family transcription factors, Gli1 to -3, which undergo extensive posttranslational modifications, including ubiquitination and SUMOylation. Here, we report that the sentrin-specific peptidase SENP1 is the specific deSUMOylation enzyme for Gli1. We show that SUMOylation stabilizes Gli1 by competing with ubiquitination at conserved lysine residues and that SUMOylated Gli1 is enriched in the nucleus, suggesting that SUMOylation is a nuclear localization signal for Gli1. Finally, we show that small interfering RNA (siRNA)-mediated knockdown of SENP1 augments the ability of Shh to sustain the proliferation of cerebellar granule cell precursors, demonstrating the physiological significance of the negative regulation of Shh signaling by SENP1.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal/fisiología , Sumoilación/fisiología , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Cerebelo/citología , Cisteína Endopeptidasas , Endopeptidasas/genética , Ratones , Ratones Noqueados , Receptor Patched-1/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Ubiquitinación/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA