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












Base de datos
Intervalo de año de publicación
1.
J Cell Biochem ; 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37260061

RESUMEN

Autophagy is a central mechanism of cellular homeostasis through the degradation of a wide range of cellular constituents. However, recent evidence suggests that autophagy actively provides information to neighboring cells via a process called secretory autophagy. Secretory autophagy couples the autophagy machinery to the secretion of cellular content via extracellular vesicles (EVs). EVs carry a variety of cargo, that reflect the pathophysiological state of the originating cells and have the potential to change the functional profile of recipient cells, to modulate cell biology. The immune system has evolved to maintain local and systemic homeostasis. It is able to sense a wide array of molecules signaling disturbed homeostasis, including EVs and their content. In this review, we explore the emerging concept of secretory autophagy as a means to communicate cellular, and in total tissue pathophysiological states to the immune system to initiate the restoration of tissue homeostasis. Understanding how autophagy mediates the secretion of immunogenic factors may hold great potential for therapeutic intervention.

2.
J Cell Biochem ; 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37126231

RESUMEN

The major histocompatibility complex (MHC) class I molecules present peptide antigens to MHC class I-restricted CD8+ T lymphocytes to elicit an effective immune response. The conventional antigen-processing pathway for MHC-I presentation depends on proteasome-mediated peptide generation and peptide loading in the endoplasmic reticulum by members of the peptide loading complex. Recent discoveries in this field highlight the role of alternative MHC-I peptide loading and presentation pathways, one of them being autophagy. Autophagy is a cell-intrinsic degradative pathway that ensures cellular homoeostasis and plays critical roles in cellular immunity. In this review article, we discuss the role of autophagy in MHC class I-restricted antigen presentation, elucidating new findings on the crosstalk of autophagy and ER-mediated MHC-I peptide presentation, dendritic cell-mediated cross-presentation and also mechanisms governing immune evasion. A detailed molecular understanding of the key drivers of autophagy-mediated MHC-I modulation holds promising targets to devise effective measures to improve T cell immunotherapies.

3.
Viruses ; 14(10)2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-36298840

RESUMEN

The human cytomegalovirus (HCMV) is a member of the beta-herpesvirus family and inflicts life-long latent infections in its hosts. HCMV has been shown to manipulate and dysregulate many cellular processes. One major interactor with the cellular host is the viral kinase pUL97. The UL97 gene is essential for viral replication, and kinase-deficient mutants of pUL97 display a severe replication defect. Recently, another group established an analog-sensitive version of the pUL97 protein. This mutant kinase can be treated with a non-hydrolysable ATP analog, thereby inhibiting its kinase function. This process is reversible by removing the ATP analog by media change. We introduced this mutant version of the pUL97 protein into the laboratory strain Ad169 of HCMV, BADwt, creating a BAD-UL97-as1 viral mutant. This mutant virus replicated normally in infected cells in the absence of the ATP analog and maintained its ability to phosphorylate its cellular substrates. However, when treated with the ATP analog, BAD-UL97-as1 displayed a defect in the production of intra- and extracellular viral DNA and in the production of viral progeny. Furthermore, in the presence of 3MB-PP1, a well-established substrate of pUL97 was no longer hyperphosphorylated. This effect was detectable as early as 4 h post treatment, which allows for studies on pUL97 without the complication of low viral titers. Nevertheless, we observed off-target effects of 3MB-PP1 on several cellular processes, which should be considered with this approach.


Asunto(s)
Citomegalovirus , ADN Viral , Humanos , Citomegalovirus/fisiología , ADN Viral/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Replicación Viral , Adenosina Trifosfato/metabolismo , Fosforilación
4.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33947818

RESUMEN

Salmonella is an intracellular pathogen of a substantial global health concern. In order to identify key players involved in Salmonella infection, we performed a global host phosphoproteome analysis subsequent to bacterial infection. Thereby, we identified the kinase SIK2 as a central component of the host defense machinery upon Salmonella infection. SIK2 depletion favors the escape of bacteria from the Salmonella-containing vacuole (SCV) and impairs Xenophagy, resulting in a hyperproliferative phenotype. Mechanistically, SIK2 associates with actin filaments under basal conditions; however, during bacterial infection, SIK2 is recruited to the SCV together with the elements of the actin polymerization machinery (Arp2/3 complex and Formins). Notably, SIK2 depletion results in a severe pathological cellular actin nucleation and polymerization defect upon Salmonella infection. We propose that SIK2 controls the formation of a protective SCV actin shield shortly after invasion and orchestrates the actin cytoskeleton architecture in its entirety to control an acute Salmonella infection after bacterial invasion.


Asunto(s)
Actinas/metabolismo , Células Epiteliales/metabolismo , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Células Epiteliales/microbiología , Células HCT116 , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Ratones , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteómica/métodos , Interferencia de ARN , Salmonella/fisiología
5.
Matrix Biol ; 100-101: 84-98, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33454423

RESUMEN

TANK-binding kinase 1 (TBK1) is a druggable multifunctional kinase that exerts a broad spectrum of functions in cells. These range from innate immunity, inflammation and interferon (IFN) signaling, through selective autophagy, specifically mitophagy and xenophagy, to energy homeostasis. Thus, it is not surprising that TBK1 is involved in many cellular signaling pathways that contribute to diverse pathologies. In this review the role of TBK1 in autophagic signaling, especially mitophagy and xenophagy, and its contribution to oncogenesis or neurodegenerative diseases will be discussed.


Asunto(s)
Autofagia , Transducción de Señal , Humanos , Inmunidad Innata , Inflamación , Proteínas Serina-Treonina Quinasas/genética
6.
EMBO Rep ; 21(1): e48317, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31709703

RESUMEN

Autophagy is a highly conserved catabolic process through which defective or otherwise harmful cellular components are targeted for degradation via the lysosomal route. Regulatory pathways, involving post-translational modifications such as phosphorylation, play a critical role in controlling this tightly orchestrated process. Here, we demonstrate that TBK1 regulates autophagy by phosphorylating autophagy modifiers LC3C and GABARAP-L2 on surface-exposed serine residues (LC3C S93 and S96; GABARAP-L2 S87 and S88). This phosphorylation event impedes their binding to the processing enzyme ATG4 by destabilizing the complex. Phosphorylated LC3C/GABARAP-L2 cannot be removed from liposomes by ATG4 and are thus protected from ATG4-mediated premature removal from nascent autophagosomes. This ensures a steady coat of lipidated LC3C/GABARAP-L2 throughout the early steps in autophagosome formation and aids in maintaining a unidirectional flow of the autophagosome to the lysosome. Taken together, we present a new regulatory mechanism of autophagy, which influences the conjugation and de-conjugation of LC3C and GABARAP-L2 to autophagosomes by TBK1-mediated phosphorylation.


Asunto(s)
Autofagosomas , Proteínas Asociadas a Microtúbulos , Autofagosomas/metabolismo , Autofagia , Familia de las Proteínas 8 Relacionadas con la Autofagia/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Péptido Hidrolasas , Fosforilación
7.
J Mol Biol ; 431(17): 3146-3156, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31247202

RESUMEN

Although the Ub-binding domain in ABIN proteins and NEMO (UBAN) is highly conserved, UBAN-containing proteins exhibit different Ub-binding properties, resulting in their diverse biological roles. Post-translational modifications further control UBAN domain specificity for poly-Ub chains. However, precisely, how the UBAN domain structurally confers such functional diversity remains poorly understood. Here we report crystal structures of ABIN-1 alone and in complex with one or two M1-linked di-Ub chains. ABIN-1 UBAN forms a homo-dimer that provides two symmetrical Ub-binding sites on either side of the coiled-coil structure. Moreover, crystal structures of ABIN1 UBAN in complex with di-Ub chains reveal a concentration-dependency of UBAN/di-Ub binding stoichiometry. Analysis of UBAN/M1-linked di-Ub binding characteristics indicates that phosphorylated S473 in OPTN and its corresponding phospho-mimetic residue in ABIN-1 (E484) are essential for high affinity interactions with M1-linked Ub chains. Also, a phospho-mimetic mutation of A303 in NEMO, corresponding to S473 of OPTN, increases binding affinity for M1-linked Ub chains. These findings are in line with the diverse physiological roles of UBAN domains, as phosphorylation of OPTN UBAN is required to enhance its binding to Ub during mitophagy.


Asunto(s)
Proteínas de Unión al ADN/química , Quinasa I-kappa B/química , Ubiquitina/química , Ubiquitina/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Humanos , Quinasa I-kappa B/genética , Mitofagia , Modelos Moleculares , Fosforilación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Análisis de Secuencia de Proteína , Ubiquitinación , Difracción de Rayos X
8.
Science ; 362(6411): 151-152, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30309930
9.
Cell Res ; 28(4): 389-390, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29572488
10.
Int J Med Microbiol ; 308(1): 176-184, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29126744

RESUMEN

Salmonella infections cause acute intestinal inflammatory responses through the action of bacterial effector proteins secreted into the host cytosol. These proteins promote Salmonella survival, amongst others, by deregulating the host innate immune system and interfering with host cell ubiquitylation signaling. This review describes the recent findings of dynamic changes of the host ubiquitinome during pathogen infection, how bacterial effector proteins modulate the host ubiquitin system and how the host innate immune system counteracts Salmonella invasion by using these pathogens as signaling platforms to initiate immune responses.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Infecciones por Salmonella/inmunología , Salmonella/metabolismo , Transducción de Señal/inmunología , Ubiquitina/metabolismo , Proteínas Bacterianas/metabolismo , Humanos , Inmunidad Innata , Salmonella/inmunología , Salmonella/patogenicidad , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
11.
Nat Microbiol ; 2: 17066, 2017 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-28481361

RESUMEN

Ubiquitination of invading Salmonella Typhimurium triggers autophagy of cytosolic bacteria and restricts their spread in epithelial cells. Ubiquitin (Ub) chains recruit autophagy receptors such as p62/SQSTM1, NDP52/CALCOCO and optineurin (OPTN), which initiate the formation of double-membrane autophagosomal structures and lysosomal destruction in a process known as xenophagy. Besides this, the functional consequences and mechanistic regulation of differentially linked Ub chains at the host-Salmonella interface have remained unexplored. Here, we show, for the first time, that distinct Ub chains on cytosolic S. Typhimurium serve as a platform triggering further signalling cascades. By using single-molecule localization microscopy, we visualized the balance and nanoscale distribution pattern of linear (M1-linked) Ub chain formation at the surface of cytosolic S. Typhimurium. In addition, we identified the deubiquitinase OTULIN as central regulator of these M1-linked Ub chains on the bacterial coat. OTULIN depletion leads to enhanced formation of linear Ub chains, resulting in local recruitment of NEMO, activation of IKKα/IKKß and ultimately NF-κB, which in turn promotes secretion of pro-inflammatory cytokines and restricts bacterial proliferation. Our results establish a role for the linear Ub coat around cytosolic S. Typhimurium as the local NF-κB signalling platform and provide insights into the function of OTULIN in NF-κB activation during bacterial pathogenesis.


Asunto(s)
Citosol/microbiología , Endopeptidasas/metabolismo , FN-kappa B/metabolismo , Salmonella typhimurium/metabolismo , Transducción de Señal , Ubiquitinación , Autofagia , Proliferación Celular , Citosol/metabolismo , Endopeptidasas/genética , Células Epiteliales/microbiología , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , FN-kappa B/genética , Salmonella typhimurium/patogenicidad , Ubiquitina/metabolismo
12.
Nat Commun ; 8: 14004, 2017 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-28084320

RESUMEN

The hallmark of Salmonella Typhimurium infection is an acute intestinal inflammatory response, which is mediated through the action of secreted bacterial effector proteins. The pro-inflammatory Salmonella effector SopA is a HECT-like E3 ligase, which was previously proposed to activate host RING ligases TRIM56 and TRIM65. Here we elucidate an inhibitory mechanism of TRIM56 and TRIM65 targeting by SopA. We present the crystal structure of SopA in complex with the RING domain of human TRIM56, revealing the atomic details of their interaction and the basis for SopA selectivity towards TRIM56 and TRIM65. Structure-guided biochemical analysis shows that SopA inhibits TRIM56 E3 ligase activity by occluding the E2-interacting surface of TRIM56. We further demonstrate that SopA ubiquitinates TRIM56 and TRIM65, resulting in their proteasomal degradation during infection. Our results provide the basis for how a bacterial HECT ligase blocks host RING ligases and exemplifies the multivalent power of bacterial effectors during infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Salmonella/enzimología , Salmonella typhimurium/enzimología , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencias de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Interacciones Huésped-Patógeno , Humanos , Proteolisis , Infecciones por Salmonella/genética , Infecciones por Salmonella/microbiología , Salmonella typhimurium/química , Salmonella typhimurium/genética , Proteínas de Motivos Tripartitos/química , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
13.
Proc Natl Acad Sci U S A ; 113(15): 4039-44, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27035970

RESUMEN

Selective autophagy of damaged mitochondria requires autophagy receptors optineurin (OPTN), NDP52 (CALCOCO2), TAX1BP1, and p62 (SQSTM1) linking ubiquitinated cargo to autophagic membranes. By using quantitative proteomics, we show that Tank-binding kinase 1 (TBK1) phosphorylates all four receptors on several autophagy-relevant sites, including the ubiquitin- and LC3-binding domains of OPTN and p62/SQSTM1 as well as the SKICH domains of NDP52 and TAX1BP1. Constitutive interaction of TBK1 with OPTN and the ability of OPTN to bind to ubiquitin chains are essential for TBK1 recruitment and kinase activation on mitochondria. TBK1 in turn phosphorylates OPTN's UBAN domain at S473, thereby expanding the binding capacity of OPTN to diverse Ub chains. In combination with phosphorylation of S177 and S513, this posttranslational modification promotes recruitment and retention of OPTN/TBK1 on ubiquitinated, damaged mitochondria. Moreover, phosphorylation of OPTN on S473 enables binding to pS65 Ub chains and is also implicated in PINK1-driven and Parkin-independent mitophagy. Thus, TBK1-mediated phosphorylation of autophagy receptors creates a signal amplification loop operating in selective autophagy of damaged mitochondria.


Asunto(s)
Autofagia , Mitocondrias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción TFIIIA/metabolismo , Proteínas de Ciclo Celular , Células HeLa , Humanos , Proteínas de Transporte de Membrana , Fosforilación , Unión Proteica , Ubiquitina/metabolismo
14.
EMBO Rep ; 16(9): 1071-83, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26268526

RESUMEN

Ubiquitylation is among the most prevalent post-translational modifications (PTMs) and regulates numerous cellular functions. Interestingly, ubiquitin (Ub) can be itself modified by other PTMs, including acetylation and phosphorylation. Acetylation of Ub on K6 and K48 represses the formation and elongation of Ub chains. Phosphorylation of Ub happens on multiple sites, S57 and S65 being the most frequently modified in yeast and mammalian cells, respectively. In mammals, the PINK1 kinase activates ubiquitin ligase Parkin by phosphorylating S65 of Ub and of the Parkin Ubl domain, which in turn promotes the amplification of autophagy signals necessary for the removal of damaged mitochondria. Similarly, TBK1 phosphorylates the autophagy receptors OPTN and p62 to initiate feedback and feedforward programs for Ub-dependent removal of protein aggregates, mitochondria and pathogens (such as Salmonella and Mycobacterium tuberculosis). The impact of PINK1-mediated phosphorylation of Ub and TBK1-dependent phosphorylation of autophagy receptors (OPTN and p62) has been recently linked to the development of Parkinson's disease and amyotrophic lateral sclerosis, respectively. Hence, the post-translational modification of Ub and its receptors can efficiently expand the Ub code and modulate its functions in health and disease.


Asunto(s)
Procesamiento Proteico-Postraduccional/genética , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinación/genética , Acetilación , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/genética , Animales , Autofagia/genética , Humanos , Mitofagia/genética , Mitofagia/fisiología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Fosforilación/genética , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Ubiquitina/química , Levaduras/genética , Levaduras/fisiología
15.
Sci Signal ; 8(372): ra35, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25872870

RESUMEN

The deubiquitylating enzyme OTUB1 is present in all tissues and targets many substrates, in both the cytosol and nucleus. We found that casein kinase 2 (CK2) phosphorylated OTUB1 at Ser(16) to promote its nuclear accumulation in cells. Pharmacological inhibition or genetic ablation of CK2 blocked the phosphorylation of OTUB1 at Ser(16), causing its nuclear exclusion in various cell types. Whereas we detected unphosphorylated OTUB1 mainly in the cytosol, we detected Ser(16)-phosphorylated OTUB1 only in the nucleus. In vitro, Ser(16)-phosphorylated OTUB1 and nonphosphorylated OTUB1 exhibited similar catalytic activity, bound K63-linked ubiquitin chains, and interacted with the E2 enzyme UBE2N. CK2-mediated phosphorylation and subsequent nuclear localization of OTUB1 promoted the formation of 53BP1 (p53-binding protein 1) DNA repair foci in the nucleus of osteosarcoma cells exposed to ionizing radiation. Our findings indicate that the activity of CK2 is necessary for the nuclear translocation and subsequent function of OTUB1 in DNA damage repair.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Núcleo Celular/metabolismo , Cisteína Endopeptidasas/metabolismo , Serina/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Animales , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/genética , Línea Celular Tumoral , Células Cultivadas , Cisteína Endopeptidasas/genética , Enzimas Desubicuitinizantes , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Lisina/genética , Lisina/metabolismo , Ratones Noqueados , Microscopía Fluorescente , Mutación , Fosforilación/efectos de los fármacos , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Serina/genética , Factor de Crecimiento Transformador beta/farmacología , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
16.
Sci Rep ; 5: 9811, 2015 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-25922883

RESUMEN

CRISPR/Cas9 technologies have been employed for genome editing to achieve gene knockouts and knock-ins in somatic cells. Similarly, certain endogenous genes have been tagged with fluorescent proteins. Often, the detection of tagged proteins requires high expression and sophisticated tools such as confocal microscopy and flow cytometry. Therefore, a simple, sensitive and robust transcriptional reporter system driven by endogenous promoter for studies into transcriptional regulation is desirable. We report a CRISPR/Cas9-based methodology for rapidly integrating a firefly luciferase gene in somatic cells under the control of endogenous promoter, using the TGFß-responsive gene PAI-1. Our strategy employed a polycistronic cassette containing a non-fused GFP protein to ensure the detection of transgene delivery and rapid isolation of positive clones. We demonstrate that firefly luciferase cDNA can be efficiently delivered downstream of the promoter of the TGFß-responsive gene PAI-1. Using chemical and genetic regulators of TGFß signalling, we show that it mimics the transcriptional regulation of endogenous PAI-1 expression. Our unique approach has the potential to expedite studies on transcription of any gene in the context of its native chromatin landscape in somatic cells, allowing for robust high-throughput chemical and genetic screens.


Asunto(s)
Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Genes Reporteros/genética , Transcripción Genética/genética , Línea Celular Tumoral , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Inhibidor 1 de Activador Plasminogénico/genética , Regiones Promotoras Genéticas/genética , Factor de Crecimiento Transformador beta/genética , Transgenes/genética , Factores de Intercambio de Guanina Nucleótido ras
17.
J Med Chem ; 58(1): 183-96, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25075558

RESUMEN

We developed a pharmacophore model for type II inhibitors that was used to guide the construction of a library of kinase inhibitors. Kinome-wide selectivity profiling of the library resulted in the identification of a series of 4-substituted 1H-pyrrolo[2,3-b]pyridines that exhibited potent inhibitory activity against two mitogen-activated protein kinases (MAPKs), TAK1 (MAP3K7) and MAP4K2, as well as pharmacologically well interrogated kinases such as p38α (MAPK14) and ABL. Further investigation of the structure-activity relationship (SAR) resulted in the identification of potent dual TAK1 and MAP4K2 inhibitors such as 1 (NG25) and 2 as well as MAP4K2 selective inhibitors such as 16 and 17. Some of these inhibitors possess good pharmacokinetic properties that will enable their use in pharmacological studies in vivo. A 2.4 Å cocrystal structure of TAK1 in complex with 1 confirms that the activation loop of TAK1 assumes the DFG-out conformation characteristic of type II inhibitors.


Asunto(s)
Quinasas Quinasa Quinasa PAM/química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Animales , Área Bajo la Curva , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Diseño de Fármacos , Descubrimiento de Drogas , Quinasas del Centro Germinal , Humanos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Modelos Químicos , Modelos Moleculares , Estructura Molecular , Fosforilación/efectos de los fármacos , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteoma/antagonistas & inhibidores , Proteoma/química , Proteoma/metabolismo , Piridinas/química , Piridinas/farmacocinética , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacocinética , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
18.
Cell Signal ; 26(10): 2186-92, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25007997

RESUMEN

The members of the transforming growth factor beta (TGFß) family of cytokines, including bone morphogenetic proteins (BMP), play fundamental roles in development and tissue homeostasis. Hence, aberrant TGFß/BMP signalling is associated with several human diseases such as fibrosis, bone and immune disorders, cancer progression and metastasis. Consequently, targeting TGFß signalling for intervention potentially offers therapeutic opportunities against these diseases. Many investigations have focussed on understanding the molecular mechanisms underpinning the regulation of TGFß signalling. One of the key areas has been to investigate the regulation of the protein components of the TGFß/BMP signal transduction pathways by ubiquitylation and deubiquitylation. In the last 15years, extensive research has led to the discovery and characterisation of several E3 ubiquitin ligases that influence the TGFß pathway. However, the research on DUBs regulating the TGFß pathway has received prominence only recently and is still an emerging field. This review will provide a concise summary of our current understanding of how DUBs regulate TGFß signalling.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Endopeptidasas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Humanos , Transducción de Señal , Proteínas Smad Reguladas por Receptores/metabolismo , Proteína Smad4/metabolismo , Proteína smad7/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación
19.
Open Biol ; 4(5): 140065, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24850914

RESUMEN

Protein kinase ALK3/BMPR1A mediates bone morphogenetic protein (BMP) signalling through phosphorylation and activation of SMADs 1/5/8. SMAD6, a transcriptional target of BMP, negatively regulates the BMP pathway by recruiting E3 ubiquitin ligases and targeting ALK3 for ubiquitin-mediated degradation. Here, we identify a deubiquitylating enzyme USP15 as an interactor of SMAD6 and ALK3. We show that USP15 enhances BMP-induced phosphorylation of SMAD1 by interacting with and deubiquitylating ALK3. RNAi-mediated depletion of USP15 increases ALK3 K48-linked polyubiquitylation, and reduces both BMP-induced SMAD1 phosphorylation and transcription of BMP target genes. We also show that loss of USP15 expression from mouse myoblast cells inhibits BMP-induced osteoblast differentiation. Furthermore, USP15 modulates BMP-induced phosphorylation of SMAD1 and transcription during Xenopus embryogenesis.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Proteína Smad1/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Ratones , Fosforilación , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Ubiquitinación
20.
Open Biol ; 4: 130210, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24554596

RESUMEN

Bone morphogenetic proteins (BMPs) control multiple cellular processes in embryos and adult tissues. BMPs signal through the activation of type I BMP receptor kinases, which then phosphorylate SMADs 1/5/8. In the canonical pathway, this triggers the association of these SMADs with SMAD4 and their translocation to the nucleus, where they regulate gene expression. BMPs can also signal independently of SMAD4, but this pathway is poorly understood. Here, we report the discovery and characterization of PAWS1/FAM83G as a novel SMAD1 interactor. PAWS1 forms a complex with SMAD1 in a SMAD4-independent manner, and BMP signalling induces the phosphorylation of PAWS1 through BMPR1A. The phosphorylation of PAWS1 in response to BMP is essential for activation of the SMAD4-independent BMP target genes NEDD9 and ASNS. Our findings identify PAWS1 as the first non-SMAD substrate for type I BMP receptor kinases and as a novel player in the BMP pathway. We also demonstrate that PAWS1 regulates the expression of several non-BMP target genes, suggesting roles for PAWS1 beyond the BMP pathway.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transducción de Señal , Proteína Smad1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Smad4/metabolismo , Especificidad por Sustrato , Factor de Crecimiento Transformador beta/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...