Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
1.
Cancer Sci ; 114(11): 4355-4364, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37688294

RESUMEN

Accumulating evidence suggests an association between iron metabolism and lung cancer progression. In biological systems, iron is present in either reduced (Fe2+ ; ferrous) or oxidized (Fe3+ ; ferric) states. However, ferrous and ferric iron exhibit distinct chemical and biological properties, the role of ferrous and ferric iron in lung cancer cell growth has not been clearly distinguished. In this study, we manipulated the balance between cellular ferrous and ferric iron status by inducing gene mutations involving the FBXL5-IRP2 axis, a ubiquitin-dependent regulatory system for cellular iron homeostasis, and determined its effects on lung cancer cell growth. FBXL5 depletion (ferrous iron accumulation) was found to suppress lung cancer cell growth, whereas IRP2 depletion (ferric iron accumulation) did not suppress such growth, suggesting that ferrous iron but not ferric iron plays a suppressive role in cell growth. Mechanistically, the depletion of FBXL5 impaired the degradation of the cyclin-dependent kinase inhibitor, p27, resulting in a delay in the cell cycle at the G1/S phase. FBXL5 depletion in lung cancer cells also improved the survival of tumor-bearing mice. Overall, this study highlights the important function of ferrous iron in cell cycle progression and lung cancer cell growth.


Asunto(s)
Proteínas F-Box , Neoplasias Pulmonares , Animales , Ratones , Complejos de Ubiquitina-Proteína Ligasa/química , Complejos de Ubiquitina-Proteína Ligasa/genética , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Neoplasias Pulmonares/genética , Hierro/metabolismo , Ubiquitina/metabolismo , Compuestos Férricos , Proteínas F-Box/genética , Proteínas F-Box/metabolismo
2.
Cancer Genomics Proteomics ; 20(3): 298-307, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37093682

RESUMEN

BACKGROUND/AIM: Alteration of F-box and leucine-rich repeat protein 5 (FBXL5), an iron-sensing ubiquitin ligase, might be related with carcinogenesis of hepatocellular carcinoma (HCC), by disturbing cellular iron homeostasis. However, the clinical implications of FBXL5 expression using patient samples need to be elucidated. PATIENTS AND METHODS: We collected HCC tissue samples from two institutes: Samsung Medical Center (n=259) and Hallym University Sacred Heart Hospital (n=115) and evaluated FBXL5 expression using immunohistochemistry. Using cut-off values determined by X-tile software, association between FBXL5 expression and several clinicopathological parameters was investigated. For external validation, the Cancer Genome Atlas (TCGA) cohort was used. RESULTS: The best cutoff value for FBXL5 IHC expression associated with recurrence-free survival (RFS) was 5%. Low FBXL5 expression was found in 18.7% of the total 374 HCCs and was associated with non-viral etiology (p=0.019). Low FBXL5 expression was related with inferior disease-specific survival (DSS, p=0.002) and RFS (p=0.001) and also was an independent prognostic factor for DSS and RFS. In addition, cases with low FBLX5 mRNA levels showed inferior DSS and RFS (p<0.001 and p=0.002, respectively) compared to high FBLX5 mRNA levels in the TCGA cohort. CONCLUSION: Down-regulation of FBXL5 expression in HCCs might be associated with poor prognosis. FBXL5 might be a prognostic biomarker of HCCs and a potential therapeutic target in conjunction with iron homeostasis.


Asunto(s)
Carcinoma Hepatocelular , Proteínas F-Box , Neoplasias Hepáticas , Humanos , Complejos de Ubiquitina-Proteína Ligasa/genética , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Proteínas Repetidas Ricas en Leucina , Hierro/metabolismo , ARN Mensajero , Pronóstico , Proteínas F-Box/genética , Proteínas F-Box/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35105802

RESUMEN

Human cytomegalovirus (HCMV) is an important human pathogen and a paradigm of viral immune evasion, targeting intrinsic, innate, and adaptive immunity. We have employed two orthogonal multiplexed tandem mass tag-based proteomic screens to identify host proteins down-regulated by viral factors expressed during the latest phases of viral infection. This approach revealed that the HIV-1 restriction factor Schlafen-11 (SLFN11) was degraded by the poorly characterized, late-expressed HCMV protein RL1, via recruitment of the Cullin4-RING E3 Ubiquitin Ligase (CRL4) complex. SLFN11 potently restricted HCMV infection, inhibiting the formation and spread of viral plaques. Overall, we show that a restriction factor previously thought only to inhibit RNA viruses additionally restricts HCMV. We define the mechanism of viral antagonism and also describe an important resource for revealing additional molecules of importance in antiviral innate immunity and viral immune evasion.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Evasión Inmune , Proteínas Nucleares/inmunología , Proteolisis , Proteínas del Envoltorio Viral/inmunología , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Humanos , Proteínas Nucleares/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Complejos de Ubiquitina-Proteína Ligasa/inmunología , Proteínas del Envoltorio Viral/genética
4.
J Am Chem Soc ; 144(2): 701-708, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-34994556

RESUMEN

Proteolysis-targeting chimeras (PROTACs), heterobifunctional compounds that consist of protein-targeting ligands linked to an E3 ligase recruiter, have arisen as a powerful therapeutic modality for targeted protein degradation (TPD). Despite the popularity of TPD approaches in drug discovery, only a small number of E3 ligase recruiters are available for the >600 E3 ligases that exist in human cells. Here, we have discovered a cysteine-reactive covalent ligand, EN106, that targets FEM1B, an E3 ligase recently discovered as the critical component of the cellular response to reductive stress. By targeting C186 in FEM1B, EN106 disrupts recognition of the key reductive stress substrate of FEM1B, FNIP1. We further establish that EN106 can be used as a covalent recruiter for FEM1B in TPD applications by demonstrating that a PROTAC linking EN106 to the BET bromodomain inhibitor JQ1 or the kinase inhibitor dasatinib leads to the degradation of BRD4 and BCR-ABL, respectively. Our study showcases a covalent ligand that targets a natural E3 ligase-substrate binding site and highlights the utility of covalent ligand screening in expanding the arsenal of E3 ligase recruiters suitable for TPD applications.


Asunto(s)
Acetamidas/química , Proteínas de Ciclo Celular/metabolismo , Proteolisis , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Animales , Azepinas/química , Sitios de Unión , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular , Cisteína/química , Dasatinib/química , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Triazoles/química , Complejos de Ubiquitina-Proteína Ligasa/antagonistas & inhibidores , Complejos de Ubiquitina-Proteína Ligasa/genética
5.
Nat Commun ; 12(1): 5779, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34599168

RESUMEN

Geminin and its binding partner Cdt1 are essential for the regulation of DNA replication. Here we show that the CULLIN3 E3 ubiquitin ligase adaptor protein SPOP binds Geminin at endogenous level and regulates DNA replication. SPOP promotes K27-linked non-degradative poly-ubiquitination of Geminin at lysine residues 100 and 127. This poly-ubiquitination of Geminin prevents DNA replication over-firing by indirectly blocking the association of Cdt1 with the MCM protein complex, an interaction required for DNA unwinding and replication. SPOP is frequently mutated in certain human cancer types and implicated in tumorigenesis. We show that cancer-associated SPOP mutations impair Geminin K27-linked poly-ubiquitination and induce replication origin over-firing and re-replication. The replication stress caused by SPOP mutations triggers replication catastrophe and cell death upon ATR inhibition. Our results reveal a tumor suppressor role of SPOP in preventing DNA replication over-firing and genome instability and suggest that SPOP-mutated tumors may be susceptible to ATR inhibitor therapy.


Asunto(s)
Geminina/metabolismo , Proteínas Nucleares/genética , Proteínas Represoras/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN/genética , Replicación del ADN/fisiología , Geminina/genética , Humanos , Masculino , Ratones , Ratones SCID , Proteínas de Mantenimiento de Minicromosoma/genética , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Mutación/genética , Ubiquitinación/genética , Ubiquitinación/fisiología
6.
Life Sci Alliance ; 4(10)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34407997

RESUMEN

Ribonucleoside monophosphate (rNMP) incorporation in genomic DNA poses a significant threat to genomic integrity. In addition to repair, DNA damage tolerance mechanisms ensure replication progression upon encountering unrepaired lesions. One player in the tolerance mechanism is Rad5, which is an E3 ubiquitin ligase and helicase. Here, we report a new role for yeast Rad5 in tolerating rNMP incorporation, in the absence of the bona fide ribonucleotide excision repair pathway via RNase H2. This role of Rad5 is further highlighted after replication stress induced by hydroxyurea or by increasing rNMP genomic burden using a mutant DNA polymerase (Pol ε - Pol2-M644G). We further demonstrate the importance of the ATPase and ubiquitin ligase domains of Rad5 in rNMP tolerance. These findings suggest a similar role for the human Rad5 homologues helicase-like transcription factor (HLTF) and SNF2 Histone Linker PHD RING Helicase (SHPRH) in rNMP tolerance, which may impact the response of cancer cells to replication stress-inducing therapeutics.


Asunto(s)
ADN Helicasas/metabolismo , Ribonucleótidos/metabolismo , Adenosina Trifosfatasas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Daño del ADN , ADN Helicasas/química , ADN Helicasas/genética , Genómica/métodos , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estrés Fisiológico , Complejos de Ubiquitina-Proteína Ligasa/genética , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Levaduras/fisiología
7.
Cells ; 10(8)2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-34440687

RESUMEN

Autoimmune liver diseases (AILD) often lead to transformation of the liver tissues into hepatocellular carcinoma (HCC). Considering the drawbacks of surgical procedures in such cases, need of successful non-invasive therapeutic strategies and treatment modalities for AILD-associated-HCC still exists. Due to the lack of clear, sufficient knowledge about factors mediating AILD-to-HCC transition, an in silico approach was adopted to delineate the underlying molecular deterministic factors. Parallel enrichment analyses on two different public microarray datasets (GSE159676 and GSE62232) pinpointed the core transcriptional regulators as key players. Correlation between the expression kinetics of these transcriptional modules in AILD and HCC was found to be positive primarily with the advancement of hepatic fibrosis. Most of the regulatory interactions were operative during early (F0-F1) and intermediate fibrotic stages (F2-F3), while the extent of activity in the regulatory network considerably diminished at late stage of fibrosis/cirrhosis (F4). Additionally, most of the transcriptional targets with higher degrees of connectivity in the regulatory network (namely DCAF11, PKM2, DGAT2 and BCAT1) may be considered as potential candidates for biomarkers or clinical targets compared to their low-connectivity counterparts. In summary, this study uncovers new possibilities in the designing of novel prognostic and therapeutic regimen for autoimmunity-associated malignancy of liver in a disease progression-dependent fashion.


Asunto(s)
Enfermedades Autoinmunes/complicaciones , Carcinoma Hepatocelular/etiología , Simulación por Computador , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/etiología , Enfermedades Autoinmunes/genética , Biomarcadores , Carcinoma Hepatocelular/genética , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Colangitis Esclerosante/complicaciones , Colangitis Esclerosante/genética , Biología Computacional , Diacilglicerol O-Acetiltransferasa/análisis , Diacilglicerol O-Acetiltransferasa/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática Biliar/complicaciones , Cirrosis Hepática Biliar/genética , Hepatopatías/complicaciones , Hepatopatías/genética , Neoplasias Hepáticas/genética , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Hormonas Tiroideas/análisis , Hormonas Tiroideas/genética , Transaminasas/análisis , Transaminasas/genética , Complejos de Ubiquitina-Proteína Ligasa/análisis , Complejos de Ubiquitina-Proteína Ligasa/genética , Proteínas de Unión a Hormona Tiroide
8.
Front Immunol ; 12: 635475, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815386

RESUMEN

Nuclear dot protein 52 kDa (NDP52, also known as CALCOCO2) functions as a selective autophagy receptor. The linear ubiquitin chain assembly complex (LUBAC) specifically generates the N-terminal Met1-linked linear ubiquitin chain, and regulates innate immune responses, such as nuclear factor-κB (NF-κB), interferon (IFN) antiviral, and apoptotic pathways. Although NDP52 and LUBAC cooperatively regulate bacterial invasion-induced xenophagy, their functional crosstalk remains enigmatic. Here we show that NDP52 suppresses canonical NF-κB signaling through the broad specificity of ubiquitin-binding at the C-terminal UBZ domain. Upon TNF-α-stimulation, NDP52 associates with LUBAC through the HOIP subunit, but does not disturb its ubiquitin ligase activity, and has a modest suppressive effect on NF-κB activation by functioning as a component of TNF-α receptor signaling complex I. NDP52 also regulates the TNF-α-induced apoptotic pathway, but not doxorubicin-induced intrinsic apoptosis. A chemical inhibitor of LUBAC (HOIPIN-8) cancelled the increased activation of the NF-κB and IFN antiviral pathways, and enhanced apoptosis in NDP52-knockout and -knockdown HeLa cells. Upon Salmonella-infection, colocalization of Salmonella, LC3, and linear ubiquitin was detected in parental HeLa cells to induce xenophagy. Treatment with HOIPIN-8 disturbed the colocalization and facilitated Salmonella expansion. In contrast, HOIPIN-8 showed little effect on the colocalization of LC3 and Salmonella in NDP52-knockout cells, suggesting that NDP52 is a weak regulator in LUBAC-mediated xenophagy. These results indicate that the crosstalk between NDP52 and LUBAC regulates innate immune responses, apoptosis, and xenophagy.


Asunto(s)
Apoptosis , Inmunidad Innata , Macroautofagia , Proteínas Nucleares/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitina/metabolismo , Células A549 , Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células HEK293 , Células HeLa , Humanos , Hidrocarburos Aromáticos/farmacología , Inmunidad Innata/efectos de los fármacos , Macroautofagia/efectos de los fármacos , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Salmonella enterica , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología , Complejos de Ubiquitina-Proteína Ligasa/antagonistas & inhibidores , Complejos de Ubiquitina-Proteína Ligasa/genética , Ubiquitinación
9.
Cancer Res Treat ; 53(4): 1174-1183, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33735560

RESUMEN

PURPOSE: Advanced stage clear cell renal cell carcinoma (ccRCC) involves a poor prognosis. Several studies have reported that dysfunctions in iron metabolism‒related proteins may cause tumor progression and metastasis of this carcinoma. In this study, we investigated the impact of the expression of iron metabolism‒related proteins on patient prognoses in advanced stage ccRCCs. MATERIALS AND METHODS: All of 143 advanced stage ccRCC specimens were selected following validation with double blind reviews. Several clinicopathological parameters including nuclear grade, perirenal fat invasion, renal sinus fat invasion, vascular invasion, necrosis, and sarcomatoid/rhabdoid differentiation were compared with the expression of ferroportin (FPN), and F-Box and leucine rich repeat protein 5 (FBXL5), by immunohistochemistry. FPN and FBXL5 mRNA level of ccRCC from The Cancer Genome Atlas database were also analyzed for validation. RESULTS: FPN and FBXL5 immunohistochemistry showed membrane and cytoplasmic expression, respectively. Based on the H-score, cases were classified as low or high expression with a cutoff value of 20 for FPN and 15 for FBXL5, respectively. Low expression of FPN and FBXL5 were significantly associated with patient death (p=0.022 and p=0.005, respectively). In survival analyses, low expression of FPN and FBXL5 were significantly associated with shorter overall survival (p=0.003 and p=0.004, respectively). On multivariate analysis, low expression of FBXL5 (hazard ratio, 2.001; p=0.034) was significantly associated with shorter overall survival. CONCLUSION: FPN and FBXL5 can be used as potential prognostic markers and therapeutic targets for advanced stage ccRCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/secundario , Proteínas de Transporte de Catión/metabolismo , Proteínas F-Box/metabolismo , Neoplasias Renales/patología , Recurrencia Local de Neoplasia/patología , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/terapia , Proteínas de Transporte de Catión/genética , Terapia Combinada , Proteínas F-Box/genética , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/terapia , Metástasis Linfática , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/terapia , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Complejos de Ubiquitina-Proteína Ligasa/genética
10.
Nat Commun ; 12(1): 734, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531470

RESUMEN

Driver genes with a mutually exclusive mutation pattern across tumor genomes are thought to have overlapping roles in tumorigenesis. In contrast, we show here that mutually exclusive prostate cancer driver alterations involving the ERG transcription factor and the ubiquitin ligase adaptor SPOP are synthetic sick. At the molecular level, the incompatible cancer pathways are driven by opposing functions in SPOP. ERG upregulates wild type SPOP to dampen androgen receptor (AR) signaling and sustain ERG activity through degradation of the bromodomain histone reader ZMYND11. Conversely, SPOP-mutant tumors stabilize ZMYND11 to repress ERG-function and enable oncogenic androgen receptor signaling. This dichotomy regulates the response to therapeutic interventions in the AR pathway. While mutant SPOP renders tumor cells susceptible to androgen deprivation therapies, ERG promotes sensitivity to high-dose androgen therapy and pharmacological inhibition of wild type SPOP. More generally, these results define a distinct class of antagonistic cancer drivers and a blueprint toward their therapeutic exploitation.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Represoras/metabolismo , Regulador Transcripcional ERG/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Ratones Desnudos , Mutación/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Neoplasias de la Próstata/genética , Unión Proteica , Proteómica , Receptores Androgénicos/metabolismo , Proteínas Represoras/genética , Transducción de Señal/fisiología , Regulador Transcripcional ERG/genética , Complejos de Ubiquitina-Proteína Ligasa/genética
11.
Prostate ; 81(1): 81-88, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33022763

RESUMEN

BACKGROUND: Recent genomic profiling has identified a subtype of prostate cancer (PCa) characterized by two key genetic alterations: missense mutation of speckle-type POZ protein (SPOP) and homozygous deletion of chromodomain helicase DNA-binding protein 1 (CHD1). Mutually exclusive with E26 transformation-specific (ETS) rearrangements, this subtype displays high genomic instability. Previous studies indicate that deficient SPOP or CHD1 alone leads to feeble prostate abnormalities and each protein is involved in DNA damage response (DDR). It remains to be determined whether CHD1 and SPOP cooperate to suppress prostate tumorigenesis and DDR. METHODS: Prostate-specific single or double knockout of Spop and Chd1 was generated with the Cre/loxP system in mice. Wild-type or mutant SPOP (F102C, F133V) overexpression and CHD1 knockdown with short hairpin RNA were created in human benign prostatic hyperplasia cell line BPH1. The levels of DNA damage and homologous recombination repair were measured by immunofluorescence staining of γH2AX and RAD51, respectively. RESULTS: Spop/Chd1 double-knockout mice displayed prostatic intraepithelial neoplasia at both young (3 months) and old (12 months) ages and failed to generate prostate adenocarcinoma. Compared with wild-type or single-knockout mice, the double-knockout prostate harbored moderately higher proliferating cells and dramatically augmented the level of γH2AX staining, although androgen receptor-positive cells and apoptotic cells remained at a similar level. In BPH1 cell line, SPOP mutant overexpression and CHD1 silencing synergistically sensitized the cells to DNA damage by camptothecin, an inducer of double-strand breaks. CONCLUSIONS: Our results indicate that SPOP and CHD1 can synergistically promote repair of naturally occurring or chemically induced DNA damages in prostate epithelial cells. Regarding the progression of the SPOP/CHD1 subtype of PCa, other functionally complementary drivers warrant further identification. The clinical implication is that this subtype of PCa may be particularly sensitive to poly(ADP-ribose) polymerase inhibitors or DNA-damaging agents.


Asunto(s)
ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Próstata/fisiología , Neoplasias de la Próstata/genética , Proteínas Represoras/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Animales , Daño del ADN , Células Epiteliales/patología , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Próstata/patología , Hiperplasia Prostática/genética , Hiperplasia Prostática/patología , Neoplasias de la Próstata/patología
12.
Cell Rep ; 32(7): 108038, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814042

RESUMEN

The 5' end of eukaryotic mRNAs is protected by the m7G-cap structure. The transcription start site nucleotide is ribose methylated (Nm) in many eukaryotes, whereas an adenosine at this position is further methylated at the N6 position (m6A) by the mammalian Phosphorylated C-terminal domain (CTD)-interacting Factor 1 (PCIF1) to generate m6Am. Here, we show that although the loss of cap-specific m6Am in mice does not affect viability or fertility, the Pcif1 mutants display reduced body weight. Transcriptome analyses of mutant mouse tissues support a role for the cap-specific m6Am modification in stabilizing transcripts. In contrast, the Drosophila Pcif1 is catalytically dead, but like its mammalian counterpart, it retains the ability to associate with the Ser5-phosphorylated CTD of RNA polymerase II (RNA Pol II). Finally, we show that the Trypanosoma Pcif1 is an m6Am methylase that contributes to the N6,N6,2'-O-trimethyladenosine (m62Am) in the hypermethylated cap4 structure of trypanosomatids. Thus, PCIF1 has evolved to function in catalytic and non-catalytic roles.


Asunto(s)
ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/genética , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Drosophila melanogaster , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transcripción Genética
13.
Clin Exp Dermatol ; 45(2): 159-164, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31323129

RESUMEN

BACKGROUND: Woodhouse-Sakati syndrome (WSS) is a rare neuroendocrine and ectodermal disorder inherited in an autosomal recessive pattern. The syndrome presents prominent clinical features, including alopecia, neuroendocrine defects, neurological findings and progressive hearing loss. The condition results from mutations in the DCAF17 gene. AIMS: To search for the underlying genetic defect in a Pakistani family with WSS phenotypes. METHODOLOGY: Whole exome sequencing was used to search for the disease-causing variant. RESULTS: Analysis of the exome data revealed a start loss sequence variant (c.1A>G, p.M1?) in DCAF17. CONCLUSION: This variant is predicted to abolish translation of the DCAF17 polypeptide. To our knowledge, this is the first start loss variant identified in the DCAF17.


Asunto(s)
Alopecia/genética , Arritmias Cardíacas/genética , Enfermedades de los Ganglios Basales/genética , Diabetes Mellitus/genética , Hipogonadismo/genética , Discapacidad Intelectual/genética , Proteínas Nucleares/genética , Eliminación de Secuencia , Complejos de Ubiquitina-Proteína Ligasa/genética , Adolescente , Adulto , Consanguinidad , Humanos , Masculino , Linaje , Fenotipo , Secuenciación del Exoma
14.
Sci Rep ; 9(1): 14354, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31586112

RESUMEN

Transcriptional factor EB (TFEB) and nuclear factor E2-related factor 2 (Nrf2) play crucial roles in the biological response against cellular stressors; however, their relationship has not yet been investigated. Here, we constructed human neuroglioma cell lines stably expressing TFEB. The expression of Nrf2-response genes, including heme oxygenase (HO)-1, glutathione-s-transferase-mu1 (GSTM1), and p62, was induced in the cell line, independent of oxidative stress. Of note, the protein level of Nrf2 was significantly increased, and its ubiquitinated fraction was reduced in stable cells compared to that in the control cells. Among E3 ubiquitin ligases known to be involved in the ubiquitination of Nrf2, DDB1 and Cullin4 associated factor 11 (DCAF11) was down-regulated at both protein and mRNA levels in stable cells, indicating that the repression of DCAF11 by TFEB may be mainly involved in the stabilization of Nrf2. In addition, the level of phosphorylated p62 at S349 was highly increased in stable cells compared to that in control cells, which could allow it to interfere with the association of Keap1 and Nrf2, thus stabilizing Nrf2. We suggest for the first time that TFEB could activate Nrf2 by increasing its stability under conditions devoid of oxidative stress.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/genética , Línea Celular Tumoral , Regulación hacia Abajo , Células HEK293 , Humanos , Fosforilación/genética , Estabilidad Proteica , Proteínas de Unión al ARN/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitinación
15.
Oncol Rep ; 42(5): 1924-1934, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31545458

RESUMEN

The present study aimed to investigate the potential mechanisms of human miR­942 in the sunitinib­resistance of renal cell carcinoma (RCC). A sunitinib­resistant OS­RC­2 cell line was established by continuous exposure to increasing concentrations of sunitinib for ~12 weeks. The expression levels of four miRNAs were determined by reverse transcription­quantitative (RT­q)PCR. miR­942 mimics were transfected into OS­RC­2 cells and RNA sequencing was performed on the miR­942­ and negative control­transfected cells. Downregulated genes, including those of long non­coding RNAs (lncRNAs) and mRNAs, were identified. The target genes of miR­942 were predicted, followed by protein­protein interaction network construction and functional enrichment analyses of miR­942 target genes. In addition, RCC RNA­seq and miRNA­seq data were downloaded from The Cancer Genome Atlas (TCGA) database. The contributions of lncRNA and/or mRNAs to survival prediction were assessed and a competing endogenous RNA (ceRNA) network consisting of miR­942, lncRNA and mRNAs was constructed. The expression levels of LINC00461, miR­942, spalt­like transcription factor 1 (SALL1), methionyl aminopeptidase 1 (METAP1) and DDB1 and CUL4 associated factor 1 (DCAF11) were verified using RT­qPCR. The role of LINC00461 in cell viability was detected by MTT assay. The expression level of miR­942 was significantly increased in sunitinib­resistant cells. A total of seven lncRNAs and 155 mRNAs were predicted as target genes of miR­942 in the miR­942 mimic­treated samples, compared with the mimic control­treated group. These potential target genes were significantly associated with 'protein binding', 'TNF­ß signaling pathway', 'negative transcriptional regulation' and 'RNA binding'. Through the integrated analysis of RNA­sequencing and TCGA data, an miR­942­related ceRNA network, which was predicted to significantly affect the survival of patients with RCC, was constructed. The expression levels of lncRNA LINC00461 and the genes SALL1, METAP1, and DCAF11 were further verified. The viability of OS­RC­2 cells was decreased following co­transfection with miR­942 mimics and LINC00641 siRNA, and was comparable to that of wild type OS­RC­2 cells (P>0.05). Therefore, lncRNA LINC00461 may act as an miR­942 ceRNA, and affect the survival of patients with RCC by regulating the expression of SALL1, METAP1 and DCAF11.


Asunto(s)
Carcinoma de Células Renales/mortalidad , Resistencia a Antineoplásicos , Neoplasias Renales/mortalidad , MicroARNs/genética , ARN Largo no Codificante/genética , Aminopeptidasas/genética , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Pronóstico , Mapas de Interacción de Proteínas , Análisis de Secuencia de ARN , Sunitinib/farmacología , Análisis de Supervivencia , Factores de Transcripción/genética , Complejos de Ubiquitina-Proteína Ligasa/genética
16.
Elife ; 82019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31294695

RESUMEN

Overexpression of anti-apoptotic proteins MCL1 and Bcl-xL are frequently observed in many cancers. Inhibitors targeting MCL1 are in clinical development, however numerous cancer models are intrinsically resistant to this approach. To discover mechanisms underlying resistance to MCL1 inhibition, we performed multiple flow-cytometry based genome-wide CRISPR screens interrogating two drugs that directly (MCL1i) or indirectly (CDK9i) target MCL1. Remarkably, both screens identified three components (CUL5, RNF7 and UBE2F) of a cullin-RING ubiquitin ligase complex (CRL5) that resensitized cells to MCL1 inhibition. We find that levels of the BH3-only pro-apoptotic proteins Bim and Noxa are proteasomally regulated by the CRL5 complex. Accumulation of Noxa caused by depletion of CRL5 components was responsible for re-sensitization to CDK9 inhibitor, but not MCL1 inhibitor. Discovery of a novel role of CRL5 in apoptosis and resistance to multiple types of anticancer agents suggests the potential to improve combination treatments.


Asunto(s)
Proteínas Cullin/genética , Quinasa 9 Dependiente de la Ciclina/genética , Neoplasias Pulmonares/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2/genética , Línea Celular Tumoral , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Ubiquitina-Proteína Ligasas/genética
17.
Cell Rep ; 27(10): 3006-3018.e4, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167144

RESUMEN

Gut mesenchyme provides key stem cell niche signals such as Wnt ligands, but how these signals are regulated is unclear. Because Hedgehog (Hh) signaling is critical for gut mesenchymal development and tumorigenesis, we investigated Hh-mediated mechanisms by analyzing mice deleted for key negative regulators of Hh signaling, Sufu and/or Spop, in the gut mesenchyme, and demonstrated their dosage-dependent roles. Although these mutants exhibit abnormal mesenchymal cell growth and functionally defective muscle layers, villification is completed with proper mesenchymal clustering, implying a permissive role for Hh signaling. These mesenchymal defects are partially rescued by Gli2 reduction. Consistent with increased epithelial proliferation caused by abnormal Hh activation in development, Sufu reduction promotes intestinal tumorigenesis, whereas Gli2 heterozygosity suppresses it. Our analyses of chromatin and GLI2 binding genomic regions reveal its transcriptional regulation of stem cell niche signals through enhancers, providing mechanistic insight into the intestinal stem cell niche in development and tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , Intestino Delgado/metabolismo , Proteínas Represoras/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Proteína Gli2 con Dedos de Zinc/metabolismo , Actinas/metabolismo , Animales , Proliferación Celular , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Intestino Delgado/crecimiento & desarrollo , Intestino Delgado/patología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Contracción Muscular , Proteínas Musculares/metabolismo , Músculos/metabolismo , Músculos/fisiología , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transducción de Señal , Nicho de Células Madre , Factor de Crecimiento Transformador beta/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/deficiencia , Complejos de Ubiquitina-Proteína Ligasa/genética , Proteínas Wnt/metabolismo , Proteína Gli2 con Dedos de Zinc/genética
18.
Eur J Med Genet ; 62(8): 103687, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31152917

RESUMEN

Woodhouse-Sakati syndrome is a rare genetic syndrome caused by homozygous mutations of the DCAF17 gene. Several endocrine organs may be affected in the course of the disease. We present a new case with pituitary iron deposition, cardiac and intestinal anomalies, with a novel mutation in DCAF17 gene. An 18-year-old female was admitted because of delayed puberty and amenorrhea. Hormonal evaluation revealed combined hyper-hypogonadotropic hypogonadism. GH and IGF-1 levels were low without short stature. ACTH levels were high and cortisol levels were supranormal with the lack of clinical findings of cortisol excess or deficiency. Pituitary MRI indicated paramagnetic substance deposition in gland. On follow-ups, non-autoimmune, insulinopenic diabetes mellitus and secondary hypothyroidism emerged. Woodhouse-Sakati syndrome was diagnosed on the basis of consistent clinical context and subsequently a novel mutation in DCAF17 was detected.


Asunto(s)
Alopecia/genética , Arritmias Cardíacas/genética , Enfermedades de los Ganglios Basales/genética , Diabetes Mellitus/genética , Hipogonadismo/genética , Discapacidad Intelectual/genética , Proteínas Nucleares/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Adolescente , Alopecia/complicaciones , Alopecia/patología , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/patología , Enfermedades de los Ganglios Basales/complicaciones , Enfermedades de los Ganglios Basales/patología , Consanguinidad , Diabetes Mellitus/patología , Femenino , Humanos , Hipogonadismo/complicaciones , Hipogonadismo/patología , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Hierro/metabolismo , Mutación/genética , Hipófisis/metabolismo
19.
Mol Cell ; 75(2): 382-393.e5, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31229404

RESUMEN

The iron-sensing protein FBXL5 is the substrate adaptor for a SKP1-CUL1-RBX1 E3 ubiquitin ligase complex that regulates the degradation of iron regulatory proteins (IRPs). Here, we describe a mechanism of FBXL5 regulation involving its interaction with the cytosolic Fe-S cluster assembly (CIA) targeting complex composed of MMS19, FAM96B, and CIAO1. We demonstrate that the CIA-targeting complex promotes the ability of FBXL5 to degrade IRPs. In addition, the FBXL5-CIA-targeting complex interaction is regulated by oxygen (O2) tension displaying a robust association in 21% O2 that is severely diminished in 1% O2 and contributes to O2-dependent regulation of IRP degradation. Together, these data identify a novel oxygen-dependent signaling axis that links IRP-dependent iron homeostasis with the Fe-S cluster assembly machinery.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Chaperonas Moleculares/genética , Complejos Multiproteicos/genética , Complejos de Ubiquitina-Proteína Ligasa/genética , Proteínas de Ciclo Celular/química , Proteínas F-Box/química , Células HeLa , Homeostasis , Humanos , Hierro/metabolismo , Proteínas Reguladoras del Hierro/genética , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/genética , Chaperonas Moleculares/química , Complejos Multiproteicos/química , Oxígeno/metabolismo , Proteolisis , Factores de Transcripción/genética , Complejos de Ubiquitina-Proteína Ligasa/química
20.
Transplantation ; 103(9): 1887-1892, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30720688

RESUMEN

BACKGROUND: Numerous studies have emphasized the genetic and phenotypic profiles of tolerant transplant patients. Moreover, different groups have defined several biomarkers, trying to distinguish patients who are going to be tolerant from those who are going to reject. However, most of these biomarkers have not been validated by other groups or even established for clinical practice. METHODS: We reanalyzed and stratified the predictive capacity of 20 previously described biomarkers for liver transplantation tolerance in a cohort of 17 liver transplant patients subjected to an independent, nonrandomized, prospective study of immunosuppression drug withdrawal. RESULTS: Only 4 of the 20 studied biomarkers (expression of SENP6, FEM1C, miR31, and miR95) showed a strong predictive capacity in the present study. miR31 and FEM1C presented an area under the ROC curve of 96.7%, followed by SENP1 with 93.3%. Finally, miR95 had an area under the ROC curve value <86.7%. CONCLUSIONS: Even though this independent analysis seems to confirm the predictive strength of SENP6 and FEM1C in liver transplantation tolerance, there are also risks in establishing biomarkers for clinical phenotypes without an understanding of how they are biologically relevant. Future collaborations between groups should be promoted so that the most promising biomarkers can be validated and implemented in daily clinical practice.


Asunto(s)
Cisteína Endopeptidasas/sangre , Supervivencia de Injerto , Trasplante de Hígado , Tolerancia al Trasplante , Complejos de Ubiquitina-Proteína Ligasa/sangre , Biomarcadores/sangre , Cisteína Endopeptidasas/genética , Rechazo de Injerto/sangre , Rechazo de Injerto/inmunología , Humanos , Trasplante de Hígado/efectos adversos , Aprendizaje Automático , Ensayos Clínicos Controlados no Aleatorios como Asunto , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Medición de Riesgo , Factores de Riesgo , Resultado del Tratamiento , Complejos de Ubiquitina-Proteína Ligasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA