Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Int J Mol Sci ; 25(12)2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38928469

RESUMEN

The SPRY domain-containing SOCS box proteins SPSB1, SPSB2, and SPSB4 utilize their SPRY/B30.2 domain to interact with a short region in the N-terminus of inducible nitric oxide synthase (iNOS), and recruit an E3 ubiquitin ligase complex to polyubiquitinate iNOS, resulting in the proteasomal degradation of iNOS. Inhibitors that can disrupt the endogenous SPSB-iNOS interactions could be used to augment cellular NO production, and may have antimicrobial and anticancer activities. We previously reported the rational design of a cyclic peptide inhibitor, cR8, cyclo(RGDINNNV), which bound to SPSB2 with moderate affinity. We, therefore, sought to develop SPSB inhibitors with higher affinity. Here, we show that cyclic peptides cR7, cyclo(RGDINNN), and cR9, cyclo(RGDINNNVE), have ~6.5-fold and ~2-fold, respectively, higher SPSB2-bindng affinities than cR8. We determined high-resolution crystal structures of the SPSB2-cR7 and SPSB2-cR9 complexes, which enabled a good understanding of the structure-activity relationships for these cyclic peptide inhibitors. Moreover, we show that these cyclic peptides displace full-length iNOS from SPSB2, SPSB1, and SPSB4, and that their inhibitory potencies correlate well with their SPSB2-binding affinities. The strongest inhibition was observed for cR7 against all three iNOS-binding SPSB proteins.


Asunto(s)
Péptidos Cíclicos , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Humanos , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/química , Oligopéptidos/química , Oligopéptidos/farmacología , Unión Proteica , Relación Estructura-Actividad
2.
Nitric Oxide ; 113-114: 1-6, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33862200

RESUMEN

Relatively high concentration of nitric oxide (NO) produced by inducible nitric oxide synthase (iNOS) in response to a variety of stimuli is a source of reactive nitrogen species, an important weapon of host innate immune defense. The SPRY domain-containing SOCS box protein 2 (SPSB2) is an E3 ubiquitin ligase that regulates the lifetime of iNOS. SPSB2 interacts with the N-terminal region of iNOS via a binding site on the SPRY domain of SPSB2, and recruits an E3 ubiquitin ligase complex to polyubiquitinate iNOS, leading to its proteasomal degradation. Although critical residues for the SPSB2-iNOS interaction have been identified, structural basis for the interaction remains to be explicitly determined. In this study, we have determined a crystal structure of the N-terminal region of iNOS in complex with the SPRY domain of SPSB2 at 1.24 Å resolution. We have resolved the roles of some flanking residues, whose contribution to the SPSB2-iNOS interaction was structurally unclear previously. Furthermore, we have evaluated the effects of SPSB2 inhibitors on NO production using transient transfection and cell-penetrating peptide approaches, and found that such inhibitors can elevate NO production in RAW264.7 macrophages. These results thus provide a useful basis for the development of potent SPSB2 inhibitors as well as recruiting ligands for proteolysis targeting chimera (PROTAC) design.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Dominio B30.2-SPRY/efectos de los fármacos , Cristalografía por Rayos X , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Modelos Moleculares , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/química , Péptidos/farmacología , Células RAW 264.7 , Proteínas Supresoras de la Señalización de Citocinas/antagonistas & inhibidores , Proteínas Supresoras de la Señalización de Citocinas/química
3.
PLoS One ; 15(11): e0242211, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33180866

RESUMEN

The IKZF1 gene, which encodes the Ikaros transcription factor, is frequently deleted or mutated in patients with B-cell precursor acute lymphoblastic leukemias that express oncogenes, like BCR-ABL, which activate the JAK-STAT5 pathway. Ikaros functionally antagonizes the transcriptional programs downstream of IL-7/STAT5 during B cell development, as well as STAT5 activity in leukemic cells. However, the mechanisms by which Ikaros interferes with STAT5 function is unknown. We studied the genomic distribution of Ikaros and STAT5 on chromatin in a murine pre-B cell line, and found that both proteins colocalize on >60% of STAT5 target regions. Strikingly, Ikaros activity leads to widespread loss of STAT5 binding at most of its genomic targets within two hours of Ikaros induction, suggesting a direct mechanism. Ikaros did not alter the level of total or phosphorylated STAT5 proteins, nor did it associate with STAT5. Using sequences from the Cish, Socs2 and Bcl6 genes that Ikaros and STAT5 target, we show that both proteins bind overlapping sequences at GGAA motifs. Our results demonstrate that Ikaros antagonizes STAT5 DNA binding, in part by competing for common target sequences. Our study has implications for understanding the functions of Ikaros and STAT5 in B cell development and transformation.


Asunto(s)
ADN/metabolismo , Factor de Transcripción Ikaros/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cromatina/metabolismo , ADN/química , Factor de Transcripción Ikaros/deficiencia , Factor de Transcripción Ikaros/genética , Interleucina-17/farmacología , Ratones , Ratones Noqueados , Fosforilación , Células Precursoras de Linfocitos B/citología , Unión Proteica , Factor de Transcripción STAT5/genética , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
4.
PLoS One ; 14(7): e0219989, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31344133

RESUMEN

Hepatitis C virus (HCV) replication involves many viral and host factors. Host factor SPRY domain- and SOCS box-containing protein 2(SPSB2) belongs to SPSB family, and it recruits target proteins by the SPRY domain and forms E3 ubiquitin ligase complexes by the SOCS box. As an adaptor protein, it can regulate the host's response to infection, but little is known about whether SPSB2 plays a role in HCV replication. In the present study, we found that HCV infection significantly upregulated the mRNA and protein levels of SPSB2 in HCVcc-infected cells. Exogenous expression of SPSB2 in hepatoma cells decreased HCV RNA and protein levels which depended on the SOCS box, while knockdown of endogenous SPSB2 increased HCV RNA and protein levels. Additionally, we demonstrated that SPSB2 interacted with HCV structural protein E1 and nonstructural protein protein 5A (NS5A) via the C-terminal portion of the SPSB2 SPRY domain. Furthermore, SPSB2 induced NS5A ubiquitination and mediated NS5A degradation. Collectively, this study discovered host factor SPSB2 significantly inhibits HCV replication by interacting and degrading NS5A.


Asunto(s)
Hepacivirus/fisiología , Hepatitis C/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Sitios de Unión , Línea Celular Tumoral , Células HEK293 , Hepatitis C/genética , Humanos , Unión Proteica , Proteolisis , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética , Ubiquitinación , Regulación hacia Arriba , Replicación Viral
5.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 6): 412-418, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-31204687

RESUMEN

The SPRY domain-containing SOCS box protein 2 (SPSB2) is one of four mammalian SPSB proteins that are characterized by a C-terminal SOCS box and a central SPRY/B30.2 domain. SPSB2 interacts with inducible nitric oxide synthase (iNOS) via the SPRY domain and polyubiquitinates iNOS, resulting in its proteasomal degradation. Inhibitors that can disrupt SPSB2-iNOS interaction and augment NO production may serve as novel anti-infective and anticancer agents. The previously determined murine SPSB2 structure may not reflect the true apo conformation of the iNOS-binding site. Here, the crystal structure of human SPSB2 SPRY domain in the apo state is reported at a resolution of 1.9 Å. Comparison of the apo and ligand-bound structures reveals that the iNOS-binding site is highly preformed and that major conformational changes do not occur upon ligand binding. Moreover, the C-terminal His6 tag of the recombinant protein binds to a shallow pocket adjacent to the iNOS-binding site on a crystallographically related SPSB2 molecule. These findings may help in structure-based and fragment-based SPSB2 inhibitor design in the future.


Asunto(s)
Apoproteínas/química , Apoproteínas/metabolismo , Modelos Moleculares , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Secuencia de Aminoácidos , Dominio B30.2-SPRY , Cristalografía por Rayos X , Humanos , Óxido Nítrico Sintasa de Tipo II/química , Conformación Proteica
6.
Cytokine ; 121: 154733, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31154249

RESUMEN

The extraordinary capacity of the liver to regenerate following injury is dependent on coordinated and regulated actions of cytokines and growth factors. Whereas hepatocyte growth factor (HGF) and epidermal growth factor (EGF) are direct mitogens to hepatocytes, inflammatory cytokines such as TNFα and IL-6 also play essential roles in the liver regeneration process. These cytokines and growth factors activate different signaling pathways in a sequential manner to elicit hepatocyte proliferation. The kinetics and magnitude of these hepatocyte-activating stimuli are tightly regulated to ensure restoration of a functional liver mass without causing uncontrolled cell proliferation. Hepatocyte proliferation can become deregulated under conditions of chronic inflammation, leading to accumulation of genetic aberrations and eventual neoplastic transformation. Among the control mechanisms that regulate hepatocyte proliferation, negative feedback inhibition by the 'suppressor of cytokine signaling (SOCS)' family proteins SOCS1 and SOCS3 play crucial roles in attenuating cytokine and growth factor signaling. Loss of SOCS1 or SOCS3 in the mouse liver increases the rate of liver regeneration and renders hepatocytes susceptible to neoplastic transformation. The frequent epigenetic repression of the SOCS1 and SOCS3 genes in hepatocellular carcinoma has stimulated research in understanding the growth regulatory mechanisms of SOCS1 and SOCS3 in hepatocytes. Whereas SOCS3 is implicated in regulating JAK-STAT signaling induced by IL-6 and attenuating EGFR signaling, SOCS1 is crucial for the regulation of HGF signaling. These two proteins also module the functions of certain key proteins that control the cell cycle. In this review, we discuss the current understanding of the functions of SOCS1 and SOCS3 in controlling hepatocyte proliferation, and its implications to liver health and disease.


Asunto(s)
Hepatocitos/citología , Hepatocitos/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Secuencia de Aminoácidos , Animales , Proliferación Celular , Transformación Celular Neoplásica/patología , Humanos , Modelos Biológicos , Proteínas Supresoras de la Señalización de Citocinas/química
7.
Nat Commun ; 10(1): 2534, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31182716

RESUMEN

The suppressor of cytokine signaling 2 (SOCS2) acts as substrate recognition subunit of a Cullin5 E3 ubiquitin ligase complex. SOCS2 binds to phosphotyrosine-modified epitopes as degrons for ubiquitination and proteasomal degradation, yet the molecular basis of substrate recognition has remained elusive. Here, we report co-crystal structures of SOCS2-ElonginB-ElonginC in complex with phosphorylated peptides from substrates growth hormone receptor (GHR-pY595) and erythropoietin receptor (EpoR-pY426) at 1.98 Å and 2.69 Å, respectively. Both peptides bind in an extended conformation recapitulating the canonical SH2 domain-pY pose, but capture different conformations of the EF loop via specific hydrophobic interactions. The flexible BG loop is fully defined in the electron density, and does not contact the substrate degron directly. Cancer-associated SNPs located around the pY pocket weaken substrate-binding affinity in biophysical assays. Our findings reveal insights into substrate recognition and specificity by SOCS2, and provide a blueprint for small molecule ligand design.


Asunto(s)
Proteínas Supresoras de la Señalización de Citocinas/química , Ubiquitina-Proteína Ligasas/química , Cristalografía por Rayos X , Humanos , Fosfotirosina/química , Polimorfismo de Nucleótido Simple , Conformación Proteica , Receptores de Eritropoyetina/química , Receptores de Somatotropina/química , Alineación de Secuencia , Especificidad por Sustrato , Proteínas Supresoras de la Señalización de Citocinas/genética , Ubiquitinación
8.
Biosci Biotechnol Biochem ; 83(2): 300-308, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30343638

RESUMEN

Suppressor of cytokine signaling (SOCS) 2, a negative regulator of growth hormone (GH) and insulin-like growth factor 1 (IGF-1), which is associated with acromegaly and cancers, is a promising candidate molecule for treating various diseases. To facilitate its use in protein therapy, we designed and constructed a human SOCS2 protein containing a membrane-permeable peptide sequence and expressed it in an Escherichia coli system. The partially purified recombinant protein was effectively delivered into several cancer cell lines and inhibited cell growth. Biochemical analysis showed that the recombinant SOCS2 protein interacted with growth hormone receptor (GHR) and downregulated GH-STAT5 signaling target genes. Our results suggest that the designed cell-penetrating SOCS2 protein will be useful in intercellular protein therapy to cure cancers. Abbreviations: SOCS: suppressor of cytokine signaling; GH: growth hormone; GHR: growth hormone receptor; IGF-1: insulin-like growth factor 1; CP: cell-penetrating; STAT: signal transducer and activator of transcription; JAK: Janus kinase; HNF: hepatocyte nuclear factor; MTM: membrane-translocating motif; HIV: human immunodeficiency virus.


Asunto(s)
Proliferación Celular/fisiología , Péptidos de Penetración Celular/metabolismo , Hormona del Crecimiento/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Escherichia coli/genética , Humanos , Janus Quinasa 2/metabolismo , Receptores de Somatotropina/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT5/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética , Transfección , Proteínas Supresoras de Tumor/metabolismo
9.
Cell Physiol Biochem ; 49(6): 2382-2395, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30261490

RESUMEN

BACKGROUND/AIMS: Glioblastoma is the most common and aggressive brain tumor and carries a poor prognosis. Previously, we found that neurotensin receptor 1 (NTSR1) contributes to glioma progression, but the underlying mechanisms of NTSR1 in glioblastoma invasion remain to be clarified. The aim of this study was to investigate the molecular mechanisms of NTSR1 in glioblastoma invasion. METHODS: Cell migration and invasion were evaluated using wound-healing and transwell assays. Cell proliferation was detected using CCK-8. The expression of NTSR1, Jun, and suppressor of cytokine signaling 6 (SOCS6) was detected using western blotting. The expression of miR-494 was detected by Quantitative real-time PCR. Chromatin immunoprecipitation assay was performed to examine the interaction between Jun and miR-494 promoter. Dual-luciferase reporter assay and western blotting were performed to identify the direct regulation of SOCS6 by miR-494. An orthotopic xenograft mouse model was conducted to assess tumor growth and invasion. RESULTS: NTSR1 knockdown attenuated the invasion of glioblastoma cells. Jun was positively regulated by NTSR1, which promoted miR-494 expression through binding to miR-494 promoter. SOCS6 was confirmed as a direct target of miR-494, thus, NTSR1-induced miR-494 upregulation resulted in SOCS6 downregulation. Both miR-494 and SOCS6 were involved in the NTSR1-induced invasion of glioblastoma cells. In vivo, tumor invasion and growth were inhibited by NTSR1 knockdown, but were restored with miR-494 overexpression. CONCLUSION: NTSR1 knockdown inhibited glioblastoma invasion via the Jun/miR-494/SOCS6 axis.


Asunto(s)
MAP Quinasa Quinasa 4/metabolismo , MicroARNs/metabolismo , Receptores de Neurotensina/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Regiones no Traducidas 3' , Animales , Antagomirs/metabolismo , Encéfalo/diagnóstico por imagen , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular , Glioblastoma/diagnóstico por imagen , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , MAP Quinasa Quinasa 4/genética , Imagen por Resonancia Magnética , Ratones , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Receptores de Neurotensina/antagonistas & inhibidores , Receptores de Neurotensina/genética , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética
10.
Sci Rep ; 8(1): 5336, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29593227

RESUMEN

Cish, participates within a multi-molecular E3 ubiquitin ligase complex, which ubiquitinates target proteins. It has an inhibitory effect on T cell activation mediated by PLC-γ1 regulation, and it functions as a potent checkpoint in CD8+ T cell tumor immunotherapy. To study the structural and functional relationships between Cish and PLC-γ1 during CD8+ T cell activation, we tested mutants of the Cish-SH2 (R107K) and D/BC (L222Q, C226Q) domains. We confirmed that Cish-SH2-specific binding was essential for PLC-γ1 ubiquitination and degradation. This domain was essential for the Cish-mediated inhibition of Ca2+ release upon TCR stimulation. No effect on inhibition of cytokine release was observed with SH2 or D/BC mutants, although the absence of Cish led to an increased release of IFN-γ and TNF-α. Using imaging we showed that Cish was expressed mostly in the cytoplasm and we did not see any Cish clustering at the plasma membrane upon stimulation. We conclude that the Cish-SH2 domain is essential for PLC-γ1 regulation in TCR-stimulated CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Fosfolipasa C gamma/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Dominios Homologos src , Animales , Linfocitos T CD8-positivos/inmunología , Calcio/metabolismo , Línea Celular , Citocinas/metabolismo , Expresión Génica , Humanos , Activación de Linfocitos , Ratones , Ratones Noqueados , Fosfolipasa C gamma/química , Fosfolipasa C gamma/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética
11.
Cell Commun Signal ; 16(1): 10, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29534718

RESUMEN

BACKGROUND: Transformation by oncogene Ras overcomes TGF-ß mediated growth inhibition in epithelial cells. However, it cooperates with each other to mediate epithelial to mesenchymal transition (EMT). The mechanism of how these two pathways interact with each other is controversial. METHODS: Molecular techniques were used to engineer expression plasmids for Ras, SPRY, TGF-ß receptors, type I and II and ubiquitin. Immunoprecipitation and western blots were employed to determine protein-protein interactions, preotein levels, protein phosphorylation while immunofluorecesent staining for molecular co-localization. TGF-ß signalling activities is also determined by its luciferase reporter assay. Trans-well assays were used to measure cell migration and invasion. RESULTS: Ras interacts with the SPSB1's SPRY domain to enhance TGF-ß signaling. Ras interacts and colocalizes with the TGF-ß type II receptor's (TßRII) negative regulator SPSB1 on the cell membrane, consequently promoting SPSB1 protein degradation via enhanced mono- and di-ubiquitination. Reduced SPSB1 levels result in the stablization of TßRII, in turn the increase of receptor levels significantly enhance Smad2/3 phosphorylation and signaling. Importantly, forced expression of SPSB1 in Ras transformed cells suppresses TGF-ß signaling and its mediated migration and invasion. CONCLUSION: Ras positively cooperates with TGF-ß signaling by reducing the cellular protein levels of TßRII negative regualtor SPSB1.


Asunto(s)
Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas ras/metabolismo , Animales , Dominio B30.2-SPRY , Movimiento Celular/efectos de los fármacos , Cicloheximida/farmacología , Perros , Regulación hacia Abajo/efectos de los fármacos , Células HEK293 , Humanos , Leupeptinas/farmacología , Células de Riñón Canino Madin Darby , Mutagénesis Sitio-Dirigida , Fosforilación/efectos de los fármacos , Unión Proteica , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Ubiquitinación , Proteínas ras/genética
12.
Fish Shellfish Immunol ; 72: 502-509, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29155031

RESUMEN

Suppressor of cytokine signaling (SOCS) is a family of cytokine-inducible negative regulators of cytokine signaling and it plays a crucial role in various physiological processes. In the present study, the full-length cDNA of a SOCS (designated as EsSOCS6) was cloned from Chinese mitten crab Eriocheir sinensis. The open reading frame of EsSOCS6 cDNA was of 1266 bp, which encoded a polypeptide of 421 amino acid residues. There were two typically conserved SOCS family domains in EsSOCS6, including a central Src homology 2 (SH2) domain and a C-terminal SOCS box. The deduced amino acid sequence of EsSOCS6 shared 72-76% similarity with those of other SOCS6 family members. EsSOCS6 mRNA was constitutively expressed in all the examined tissues with higher expression levels in the immune-related tissues, such as hepatopancreas, hemocytes and gill. The mRNA expression levels of the EsSOCS6 in hemocytes were significantly up-regulated after the stimulations with lipopolysaccharide (LPS), Aeromonas hydrophila and polyinosinic-polycytidylic acid (poly (I:C)). The mRNA expressions of threonine/serine protein kinase (EsAkt) and EsRelish were dramatically declined after EsSOCS6 was interfered by dsRNA. Collectively, these results demonstrated that EsSOCS6 might regulate the activation of the NF-κB signaling pathway and play an important role in the innate immune responses of E. sinensis.


Asunto(s)
Braquiuros/genética , Braquiuros/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Aeromonas hydrophila/fisiología , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Perfilación de la Expresión Génica , Lipopolisacáridos/farmacología , Filogenia , Poli I-C/farmacología , Alineación de Secuencia , Proteínas Supresoras de la Señalización de Citocinas/química
13.
Biochem Biophys Res Commun ; 489(3): 346-352, 2017 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-28549582

RESUMEN

SPRY domain-containing SOCS box protein 2 (SPSB2) is a negative regulator of inducible nitric oxide synthase (iNOS) that modulates the lifetime of iNOS and thus the levels of nitric oxide (NO) production. Inhibitors that can disrupt the endogenous SPSB2-iNOS interaction and augment NO production have potential as novel antimicrobial and anticancer drugs. In this study, we have designed a cyclic peptide (cR8), containing an RGD motif and the SPSB2 binding motif (DINNNV). ITC and chemical shift perturbation showed that cR8 binds to the iNOS binding site on SPSB2 with a Kd of 671 nM, and saturation transfer difference NMR showed that cR8 binds to αvß3 integrin-expressing cells. Moreover, we determined the crystal structure of SPSB2 in complex with cR8, at a resolution of 1.34 Å. cR8 forms extensive hydrogen bonding with SPSB2 residues, but loss of an intramolecular hydrogen bond that is present in SPSB2-bound iNOS peptide may destabilize the bound conformation of cR8 and lead to a gentle reduction in SPSB2 binding affinity. These results serve as a useful basis for designing site-directed SPSB2 inhibitors in the future.


Asunto(s)
Diseño de Fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oligopéptidos/farmacología , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Cristalización , Cristalografía , Humanos , Modelos Moleculares , Conformación Molecular , Oligopéptidos/química , Péptidos Cíclicos/síntesis química , Unión Proteica/efectos de los fármacos , Relación Estructura-Actividad
14.
Biochemistry ; 56(19): 2455-2466, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28437072

RESUMEN

Grafting bioactive peptide sequences onto small cysteine-rich scaffolds is a promising strategy for enhancing their stability and value as novel peptide-based therapeutics. However, correctly folded disulfide-rich peptides can be challenging to produce by either recombinant or synthetic means. The single disulfide-directed ß-hairpin (SDH) fold, first observed in contryphan-Vc1, provides a potential alternative to complex disulfide-rich scaffolds. We have undertaken recombinant production of full-length contryphan-Vc1 (rCon-Vc1[Z1Q]) and a truncated analogue (rCon-Vc11-22[Z1Q]), analyzed the backbone dynamics of rCon-Vc1[Z1Q], and probed the conformational and proteolytic stability of these peptides to evaluate the potential of contryphan-Vc1 as a molecular scaffold. Backbone 15N relaxation measurements for rCon-Vc1[Z1Q] indicate that the N-terminal domain of the peptide is ordered up to Thr19, whereas the remainder of the C-terminal region is highly flexible. The solution structure of truncated rCon-Vc11-22[Z1Q] was similar to that of the full-length peptide, indicating that the flexible C-terminus does not have any effect on the structured domain of the peptide. Contryphan-Vc1 exhibited excellent proteolytic stability against trypsin and chymotrypsin but was susceptible to pepsin digestion. We have investigated whether contryphan-Vc1 can accept a bioactive epitope while maintaining the structure of the peptide by introducing peptide sequences based on the DINNN motif of inducible nitric oxide synthase. We show that sCon-Vc11-22[NNN12-14] binds to the iNOS-binding protein SPSB2 with an affinity of 1.3 µM while maintaining the SDH fold. This study serves as a starting point in utilizing the SDH fold as a peptide scaffold.


Asunto(s)
Conotoxinas/química , Péptidos Cíclicos/química , Ingeniería de Proteínas , Proteínas Supresoras de la Señalización de Citocinas/química , Conotoxinas/genética , Conotoxinas/metabolismo , Cisteína/química , Cistina/química , Epítopos , Humanos , Cinética , Isótopos de Nitrógeno , Oxidación-Reducción , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Péptidos Cíclicos/genética , Péptidos Cíclicos/metabolismo , Conformación Proteica en Lámina beta , Pliegue de Proteína , Estabilidad Proteica , Proteolisis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Solubilidad , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Resonancia por Plasmón de Superficie
15.
Cell Signal ; 31: 41-57, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28038963

RESUMEN

The JAK2 V617F mutant-mediated aberrant signaling pathway is a hallmark of myeloproliferative neoplasms (MPNs). Although cytokine-inducible Src homology 2 protein (CIS) and suppressors of cytokine signaling (SOCS) are negative regulators of the JAK-STAT pathway, the functional role of CIS/SOCS family members in the JAK2 V617F mutant-induced oncogenic signaling pathway has not yet been elucidated. In this study, we found that the expression of CIS and SOCS1 was induced through the activation of signal transducer and activator of transcription 5 (STAT5) in not only the cells stimulated with Epo or IL-3 but also the cells transformed by the JAK2 V617F mutant. Cell proliferation and tumor formation in nude mice induced by the JAK2 V617F mutant were significantly enhanced when the expression of CIS was silenced using an RNA interference technique, whereas the knockdown of SOCS1 had no effect. The enforced expression of CIS caused apoptotic cell death in the transformed by JAK2 V617F mutant and drastically inhibited the JAK2 V617F mutant-induced tumor formation. CIS interacted with phosphorylated EpoR at Y401, which was critical for the activation of STAT5 and ERK. Whereas the activation of STAT5 and ERK in the transformed cells by JAK2 V617F mutant was increased by the knockdown of CIS, the enforced expression of CIS reduced the activation of these molecules. Furthermore, these anti-tumor effects of CIS required the function of SH2 domain and its tyrosine phosphorylation at Y253. We herein elucidated the mechanism by which CIS functions as a novel type of tumor suppressor in JAK2 V617F mutant-induced tumorigenesis.


Asunto(s)
Eritropoyetina/metabolismo , Janus Quinasa 2/metabolismo , Mutación/genética , Receptores de Eritropoyetina/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Secuencia de Aminoácidos , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Fosforilación , Unión Proteica , Factor de Transcripción STAT5/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/química
16.
Biochemistry ; 54(30): 4672-82, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26173083

RESUMEN

SOCS5 can negatively regulate both JAK/STAT and EGF-receptor pathways and has therefore been implicated in regulating both the immune response and tumorigenesis. Understanding the molecular basis for SOCS5 activity may reveal novel ways to target key components of these signaling pathways. The N-terminal region of SOCS5 coordinates critical protein interactions involved in inhibition of JAK/STAT signaling, and a conserved region within the N-terminus of SOCS5 mediates direct binding to the JAK kinase domain. Here we have characterized the solution conformation of this conserved JAK interaction region (JIR) within the largely disordered N-terminus of SOCS5. Using nuclear magnetic resonance (NMR) chemical shift analysis, relaxation measurements, and NOE analysis, we demonstrate the presence of preformed structural elements in the JIR of mouse SOCS5 (mSOCS5175-244), consisting of an α-helix encompassing residues 224-233, preceded by a turn and an extended structure. We have identified a phosphorylation site (Ser211) within the JIR of mSOCS5 and have investigated the role of phosphorylation in modulating JAK binding using site-directed mutagenesis.


Asunto(s)
Proteínas Supresoras de la Señalización de Citocinas/química , Sustitución de Aminoácidos , Animales , Ratones , Mutagénesis Sitio-Dirigida , Mutación Missense , Fosforilación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
17.
PLoS One ; 10(6): e0131218, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121586

RESUMEN

Suppressor of cytokine signalling 2 (SOCS2) is the substrate-binding component of a Cullin-RING E3 ubiquitin ligase (CRL) complex that targets phosphorylated hormone receptors for degradation by the ubiquitin-proteasome system. As a key regulator of the transcriptional response to growth signals, SOCS2 and its protein complex partners are potential targets for small molecule development. We found that crystals of SOCS2 in complex with its adaptor proteins, Elongin C and Elongin B, underwent a change in crystallographic parameters when treated with dimethyl sulfoxide during soaking experiments. To solve the phase problem for the new crystal form we identified the presence of arsenic atoms in the crystals, a result of covalent modification of cysteines by cacodylate, and successfully extracted anomalous signal from these atoms for experimental phasing. The resulting structure provides a means for solving future structures where the crystals must be treated with DMSO for ligand soaking approaches. Additionally, the conformational changes induced in this structure reveal flexibility within SOCS2 that match those postulated by previous molecular dynamics simulations. This conformational flexibility illustrates how SOCS2 can orient its substrates for successful ubiquitination by other elements of the CRL complex.


Asunto(s)
Arsenicales/química , Ácido Cacodílico/química , Proteínas Cullin/química , Dimetilsulfóxido/química , Proteínas Supresoras de la Señalización de Citocinas/química , Factores de Transcripción/química , Arsenicales/metabolismo , Ácido Cacodílico/metabolismo , Cristalografía por Rayos X , Proteínas Cullin/metabolismo , Cisteína/metabolismo , Elonguina , Humanos , Modelos Moleculares , Conformación Proteica , Soluciones , Especificidad por Sustrato , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factores de Transcripción/metabolismo
18.
J Autoimmun ; 62: 31-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26094775

RESUMEN

Uveitis is a diverse group of potentially sight-threatening intraocular inflammatory diseases and pathology derives from sustained production of pro-inflammatory cytokines in the optical axis. Although topical or systemic steroids are effective therapies, their adverse effects preclude prolonged usage and are impetus for seeking alternative immunosuppressive agents, particularly for patients with refractory uveitis. In this study, we synthesized a 16 amino acid membrane-penetrating lipophilic suppressor of cytokine signaling 1 peptide (SOCS1-KIR) that inhibits JAK/STAT signaling pathways and show that it suppresses and ameliorates experimental autoimmune uveitis (EAU), the mouse model of human uveitis. Fundus images, histological and optical coherence tomography analysis of eyes showed significant suppression of clinical disease, with average clinical score of 0.5 compared to 2.0 observed in control mice treated with scrambled peptide. We further show that SOCS1-KIR conferred protection from ocular pathology by inhibiting the expansion of pathogenic Th17 cells and inhibiting trafficking of inflammatory cells into the neuroretina during EAU. Dark-adapted scotopic and photopic electroretinograms further reveal that SOCS1-KIR prevented decrement of retinal function, underscoring potential neuroprotective effects of SOCS1-KIR in uveitis. Importantly, SOCS1-KIR is non-toxic, suggesting that topical administration of SOCS1-Mimetics can be exploited as a non-invasive treatment for uveitis and for limiting cytokine-mediated pathology in other ocular inflammatory diseases including scleritis.


Asunto(s)
Antiinflamatorios/administración & dosificación , Péptidos/administración & dosificación , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Uveítis/inmunología , Uveítis/metabolismo , Administración Tópica , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Angiografía con Fluoresceína , Inmunidad , Ratones , Retina/inmunología , Retina/metabolismo , Retina/patología , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/química , Linfocitos T/inmunología , Linfocitos T/metabolismo , Uveítis/tratamiento farmacológico , Uveítis/patología
19.
Cell Physiol Biochem ; 35(1): 227-36, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25591765

RESUMEN

BACKGROUND/AIMS: Colorectal cancer (CRC) is one of the most common malignancies worldwide, and microRNAs play a crucial role in CRC biology. The purpose of this study was to investigate the exact functions and potential mechanisms of action of miR-301a in CRC. METHODS: Quantitative real-time PCR was conducted to assess the expression of miR-301a. Cell proliferation was detected using MTT and colony formation assay, and cell invasion and migration were evaluated using Transwell assay. Luciferase reporter assay was used to identify the direct regulation of suppressor of cytokine signaling 6 (SOCS6) by miR-301a. RESULTS: We first confirmed the upregulation of miR-301a in CRC tissues and cell lines. Gain-of-function and loss-of-function studies in the human CRC cell lines, SW480 and SW620, showed that miR-301a acts as an oncogene by increasing cell proliferation, migration and invasion as well as tumor growth. Furthermore, SOCS6 was identified as a target gene of miR-301a. Reintroduction of SOCS6 partially abrogated miR-301a-induced cell proliferation, migration and invasion. CONCLUSION: These data suggest that miR-301a promotes CRC progression by directly downregulating SOCS6 expression, and miR-301a may represent a novel biomarker for the prevention and treatment of CRC.


Asunto(s)
Neoplasias Colorrectales/patología , MicroARNs/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , Oligonucleótidos Antisentido/metabolismo , ARN Mensajero/metabolismo , Alineación de Secuencia , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/genética , Trasplante Heterólogo , Regulación hacia Arriba
20.
Cancer Biol Ther ; 16(1): 160-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25561270

RESUMEN

Several microRNAs (miRNA) have been implicated in H. pylori related gastric cancer (GC). However, the molecular mechanism of miRNAs in gastric cancer has not been fully understood. In this study, we reported that miR-101 is significantly down-regulated in H. pylori positive tissues and cells and in tumor tissues with important functional consequences. Ectopic expression of miR-101 dramatically suppressed cell proliferation and colony formation by inducing G1-phase cell-cycle arrest. We found that miR-101 strongly reduced the expression of SOCS2 oncogene in GC cells. Similar to the restoring miR-26 expression, SOCS2 down-regulation inhibited cell growth and cell-cycle progression, whereas SOCS2 over-expression rescued the suppressive effect of miR-101. Mechanistic investigations revealed that miR-101 suppressed the expression of c-myc, CDK2, CDK4, CDK6, CCND2, CCND3, and CCNE2, while promoted tumor suppressor p14, p16, p21 and p27 expression. In clinical specimens, SOCS2 was over-expressed in tumors and H. pylori positive tissues and its mRNA levels were inversely correlated with miR-101 expression. Taken together, our results indicated that miR-101 functions as a growth-suppressive miRNA in H. pylori related GC, and that its suppressive effects are mediated mainly by repressing SOCS2 expression.


Asunto(s)
Transformación Celular Neoplásica/genética , Infecciones por Helicobacter/complicaciones , Helicobacter pylori , MicroARNs/genética , Neoplasias Gástricas/etiología , Neoplasias Gástricas/patología , Proteínas Supresoras de la Señalización de Citocinas/genética , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Sitios de Unión , Ciclo Celular/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , MicroARNs/química , Interferencia de ARN , Proteínas Supresoras de la Señalización de Citocinas/química , Carga Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA