Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell ; 180(6): 1245-1261.e21, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32142654

RESUMEN

In response to transcription-blocking DNA damage, cells orchestrate a multi-pronged reaction, involving transcription-coupled DNA repair, degradation of RNA polymerase II (RNAPII), and genome-wide transcription shutdown. Here, we provide insight into how these responses are connected by the finding that ubiquitylation of RNAPII itself, at a single lysine (RPB1 K1268), is the focal point for DNA-damage-response coordination. K1268 ubiquitylation affects DNA repair and signals RNAPII degradation, essential for surviving genotoxic insult. RNAPII degradation results in a shutdown of transcriptional initiation, in the absence of which cells display dramatic transcriptome alterations. Additionally, regulation of RNAPII stability is central to transcription recovery-persistent RNAPII depletion underlies the failure of this process in Cockayne syndrome B cells. These data expose regulation of global RNAPII levels as integral to the cellular DNA-damage response and open the intriguing possibility that RNAPII pool size generally affects cell-specific transcription programs in genome instability disorders and even normal cells.


Asunto(s)
Daño del ADN , ARN Polimerasa II/metabolismo , Reparación del ADN , Células HEK293 , Humanos , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transcripción Genética , Ubiquitinación , Rayos Ultravioleta
2.
Mol Cell ; 79(2): 332-341.e7, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32521225

RESUMEN

The Ddi1/DDI2 proteins are ubiquitin shuttling factors, implicated in a variety of cellular functions. In addition to ubiquitin-binding and ubiquitin-like domains, they contain a conserved region with similarity to retroviral proteases, but whether and how DDI2 functions as a protease has remained unknown. Here, we show that DDI2 knockout cells are sensitive to proteasome inhibition and accumulate high-molecular weight, ubiquitylated proteins that are poorly degraded by the proteasome. These proteins are targets for the protease activity of purified DDI2. No evidence for DDI2 acting as a de-ubiquitylating enzyme was uncovered, which could suggest that it cleaves the ubiquitylated protein itself. In support of this idea, cleavage of transcription factor NRF1 is known to require DDI2 activity in vivo. We show that DDI2 is indeed capable of cleaving NRF1 in vitro but only when NRF1 protein is highly poly-ubiquitylated. Together, these data suggest that DDI2 is a ubiquitin-directed endoprotease.


Asunto(s)
Proteasas de Ácido Aspártico/metabolismo , Factor Nuclear 1 de Respiración/metabolismo , Ubiquitina/metabolismo , Proteasas de Ácido Aspártico/genética , Sitios de Unión , Sistemas CRISPR-Cas , Línea Celular , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Biosíntesis de Proteínas , Proteolisis
3.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33883278

RESUMEN

Cancer cells can survive chemotherapy-induced stress, but how they recover from it is not known. Using a temporal multiomics approach, we delineate the global mechanisms of proteotoxic stress resolution in multiple myeloma cells recovering from proteasome inhibition. Our observations define layered and protracted programs for stress resolution that encompass extensive changes across the transcriptome, proteome, and metabolome. Cellular recovery from proteasome inhibition involved protracted and dynamic changes of glucose and lipid metabolism and suppression of mitochondrial function. We demonstrate that recovering cells are more vulnerable to specific insults than acutely stressed cells and identify the general control nonderepressable 2 (GCN2)-driven cellular response to amino acid scarcity as a key recovery-associated vulnerability. Using a transcriptome analysis pipeline, we further show that GCN2 is also a stress-independent bona fide target in transcriptional signature-defined subsets of solid cancers that share molecular characteristics. Thus, identifying cellular trade-offs tied to the resolution of chemotherapy-induced stress in tumor cells may reveal new therapeutic targets and routes for cancer therapy optimization.


Asunto(s)
Neoplasias/tratamiento farmacológico , Estrés Fisiológico/efectos de los fármacos , Antineoplásicos/farmacología , Autofagia/fisiología , Línea Celular Tumoral , Humanos , Metaboloma/genética , Mitocondrias/metabolismo , Mieloma Múltiple/metabolismo , Neoplasias/metabolismo , Neoplasias/fisiopatología , Inhibidores de Proteasoma/farmacología , Proteolisis , Proteoma/genética , Análisis de Sistemas , Transcriptoma/genética
4.
Artículo en Inglés | MEDLINE | ID: mdl-38653840

RESUMEN

While monospecific antibodies have long been the foundational offering of protein therapeutics, recent advancements in antibody engineering have allowed for the development of far more complex antibody structures. Novel molecular format (NMF) proteins, such as bispecific antibodies (BsAbs), are structures capable of multispecific binding, allowing for expanded therapeutic functionality. As demand for NMF proteins continues to rise, biomanufacturers face the challenge of increasing bioreactor process productivity while simultaneously maintaining consistent product quality. This challenge is exacerbated when producing structurally complex proteins with asymmetric modalities, as seen in NMFs. In this study, the impact of a high inoculation density (HID) fed-batch process on the productivity and product quality attributes of two CHO cell lines expressing unique NMFs, a monospecific antibody with an Fc-fusion protein and a bispecific antibody, compared to low inoculation density (LID) platform fed-batch processes was evaluated. It was observed that an intensified platform fed-batch process increased product concentrations by 33 and 109% for the two uniquely structured complex proteins in a shorter culture duration while maintaining similar product quality attributes to traditional fed-batch processes.

5.
J Biol Chem ; 297(5): 101294, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34634303

RESUMEN

Tandem mass spectrometry (MS/MS) is an accurate tool to assess modified ribonucleosides and their dynamics in mammalian cells. However, MS/MS quantification of lowly abundant modifications in non-ribosomal RNAs is unreliable, and the dynamic features of various modifications are poorly understood. Here, we developed a 13C labeling approach, called 13C-dynamods, to quantify the turnover of base modifications in newly transcribed RNA. This turnover-based approach helped to resolve mRNA from ncRNA modifications in purified RNA or free ribonucleoside samples and showed the distinct kinetics of the N6-methyladenosine (m6A) versus 7-methylguanosine (m7G) modification in polyA+-purified RNA. We uncovered that N6,N6-dimethyladenosine (m62A) exhibits distinct turnover in small RNAs and free ribonucleosides when compared to known m62A-modified large rRNAs. Finally, combined measurements of turnover and abundance of these modifications informed on the transcriptional versus posttranscriptional sensitivity of modified ncRNAs and mRNAs, respectively, to stress conditions. Thus, 13C-dynamods enables studies of the origin of modified RNAs at steady-state and subsequent dynamics under nonstationary conditions. These results open new directions to probe the presence and biological regulation of modifications in particular RNAs.


Asunto(s)
Adenosina , Isótopos de Carbono , Guanosina/análogos & derivados , Procesamiento Postranscripcional del ARN , ARN , Adenosina/química , Adenosina/metabolismo , Adenosina/farmacología , Isótopos de Carbono/química , Isótopos de Carbono/farmacología , Guanosina/química , Guanosina/metabolismo , Guanosina/farmacología , Marcaje Isotópico , ARN/química , ARN/metabolismo , Espectrometría de Masas en Tándem
6.
PLoS Pathog ; 16(6): e1008640, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32569299

RESUMEN

Ubiquitylation is a common post translational modification of eukaryotic proteins and in the human malaria parasite, Plasmodium falciparum (Pf) overall ubiquitylation increases in the transition from intracellular schizont to extracellular merozoite stages in the asexual blood stage cycle. Here, we identify specific ubiquitylation sites of protein substrates in three intraerythrocytic parasite stages and extracellular merozoites; a total of 1464 sites in 546 proteins were identified (data available via ProteomeXchange with identifier PXD014998). 469 ubiquitylated proteins were identified in merozoites compared with only 160 in the preceding intracellular schizont stage, suggesting a large increase in protein ubiquitylation associated with merozoite maturation. Following merozoite invasion of erythrocytes, few ubiquitylated proteins were detected in the first intracellular ring stage but as parasites matured through trophozoite to schizont stages the apparent extent of ubiquitylation increased. We identified commonly used ubiquitylation motifs and groups of ubiquitylated proteins in specific areas of cellular function, for example merozoite pellicle proteins involved in erythrocyte invasion, exported proteins, and histones. To investigate the importance of ubiquitylation we screened ubiquitin pathway inhibitors in a parasite growth assay and identified the ubiquitin activating enzyme (UBA1 or E1) inhibitor MLN7243 (TAK-243) to be particularly effective. This small molecule was shown to be a potent inhibitor of recombinant PfUBA1, and a structural homology model of MLN7243 bound to the parasite enzyme highlights avenues for the development of P. falciparum specific inhibitors. We created a genetically modified parasite with a rapamycin-inducible functional deletion of uba1; addition of either MLN7243 or rapamycin to the recombinant parasite line resulted in the same phenotype, with parasite development blocked at the schizont stage. Nuclear division and formation of intracellular structures was interrupted. These results indicate that the intracellular target of MLN7243 is UBA1, and this activity is essential for the final differentiation of schizonts to merozoites.


Asunto(s)
Merozoítos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Humanos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Ubiquitina/genética
7.
Methods ; 159-160: 146-156, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30769100

RESUMEN

Transcribing RNA polymerase II (RNAPII) is decorated by a plethora of post-translational modifications that mark different stages of transcription. One important modification is RNAPII ubiquitylation, which occurs in response to numerous different stimuli that cause RNAPII stalling, such as DNA damaging agents, RNAPII inhibitors, or depletion of the nucleotide pool. Stalled RNAPII triggers a so-called "last resort pathway", which involves RNAPII poly-ubiquitylation and proteasome-mediated degradation. Different approaches have been described to study RNAPII poly-ubiquitylation and degradation, each method with its own advantages and caveats. Here, we describe optimised strategies for detecting ubiquitylated RNAPII and studying its degradation, but these protocols are suitable for studying other ubiquitylated proteins as well.


Asunto(s)
ARN Polimerasa II/análisis , ARN Polimerasa II/metabolismo , Ubiquitinación , Animales , Daño del ADN , Humanos , Mamíferos/genética , Mamíferos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , ARN Polimerasa II/antagonistas & inhibidores , ARN Polimerasa II/genética , Transcripción Genética , Rayos Ultravioleta , Levaduras/enzimología , Levaduras/genética , Levaduras/metabolismo
8.
Mol Cell Proteomics ; 15(2): 542-57, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26685126

RESUMEN

Neurons require efficient transport mechanisms such as fast axonal transport to ensure neuronal homeostasis and survival. Neurotrophins and their receptors are conveyed via fast axonal retrograde transport of signaling endosomes to the soma, where they elicit transcriptional responses. Despite the essential roles of signaling endosomes in neuronal differentiation and survival, little is known about their molecular identity, dynamics, and regulation. Gaining a better mechanistic understanding of these organelles and their kinetics is crucial, given the growing evidence linking vesicular trafficking deficits to neurodegeneration. Here, we exploited an affinity purification strategy using the binding fragment of tetanus neurotoxin (HCT) conjugated to monocrystalline iron oxide nanoparticles (MIONs), which in motor neurons, is transported in the same carriers as neurotrophins and their receptors. To quantitatively assess the molecular composition of HCT-containing signaling endosomes, we have developed a protocol for triple Stable Isotope Labeling with Amino acids in Cell culture (SILAC) in embryonic stem cell-derived motor neurons. After HCT internalization, retrograde carriers were magnetically isolated at different time points and subjected to mass-spectrometry and Gene Ontology analyses. This purification strategy is highly specific, as confirmed by the presence of essential regulators of fast axonal transport in the make-up of these organelles. Our results indicate that signaling endosomes undergo a rapid maturation with the acquisition of late endosome markers following a specific time-dependent kinetics. Strikingly, signaling endosomes are specifically enriched in proteins known to be involved in neurodegenerative diseases and neuroinfection. Moreover, we highlighted the presence of novel components, whose precise temporal recruitment on signaling endosomes might be essential for proper sorting and/or transport of these organelles. This study provides the first quantitative proteomic analysis of signaling endosomes isolated from motor neurons and allows the assembly of a functional map of these axonal carriers involved in long-range neuronal signaling.


Asunto(s)
Axones/metabolismo , Neuronas Motoras/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Enfermedades Neurodegenerativas/genética , Proteómica , Animales , Transporte Axonal/efectos de los fármacos , Transporte Axonal/genética , Axones/efectos de los fármacos , Axones/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Endocitosis/genética , Endosomas/genética , Endosomas/metabolismo , Endosomas/patología , Compuestos Férricos/administración & dosificación , Compuestos Férricos/química , Humanos , Marcaje Isotópico , Nanopartículas del Metal/administración & dosificación , Nanopartículas del Metal/química , Metaloendopeptidasas/administración & dosificación , Metaloendopeptidasas/química , Ratones , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , Factores de Crecimiento Nervioso/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Transducción de Señal , Toxina Tetánica/administración & dosificación , Toxina Tetánica/química
9.
Nucleic Acids Res ; 44(11): 5246-55, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-27060134

RESUMEN

Cockayne syndrome B (CSB), best known for its role in transcription-coupled nucleotide excision repair (TC-NER), contains a ubiquitin-binding domain (UBD), but the functional connection between protein ubiquitylation and this UBD remains unclear. Here, we show that CSB is regulated via site-specific ubiquitylation. Mass spectrometry analysis of CSB identified lysine (K) 991 as a ubiquitylation site. Intriguingly, mutation of this residue (K991R) does not affect CSB's catalytic activity or protein stability, but greatly affects genome stability, even in the absence of induced DNA damage. Moreover, cells expressing CSB K991R are sensitive to oxidative DNA damage, but proficient for TC-NER. K991 becomes ubiquitylated upon oxidative DNA damage, and while CSB K991R is recruited normally to such damage, it fails to dissociate in a timely manner, suggesting a requirement for K991 ubiquitylation in CSB activation. Interestingly, deletion of CSB's UBD gives rise to oxidative damage sensitivity as well, while CSB ΔUBD and CSB K991R affects expression of overlapping groups of genes, further indicating a functional connection. Together, these results shed new light on the regulation of CSB, with K991R representing an important separation-of-function-mutation in this multi-functional protein.


Asunto(s)
Síndrome de Cockayne/genética , Síndrome de Cockayne/metabolismo , Daño del ADN , Reparación del ADN , Estrés Oxidativo , Transcripción Genética , Secuencia de Aminoácidos , Ciclo Celular , Línea Celular , Supervivencia Celular , Análisis por Conglomerados , Daño del ADN/efectos de la radiación , Expresión Génica , Perfilación de la Expresión Génica , Inestabilidad Genómica , Humanos , Mutación , Proteínas Recombinantes de Fusión , Ubiquitinación
10.
mSphere ; 8(6): e0051123, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-37975677

RESUMEN

IMPORTANCE: Toxoplasma gondii (Tg) is a ubiquitous parasitic pathogen, infecting about one-third of the global population. Tg is controlled in immunocompetent people by mechanisms that are not fully understood. Tg infection drives the production of the inflammatory cytokine interferon gamma (IFNγ), which upregulates intracellular anti-pathogen defense pathways. In this study, we describe host proteins p97/VCP, UBXD1, and ANKRD13A that control Tg at the parasitophorous vacuole (PV) in IFNγ-stimulated endothelial cells. p97/VCP is an ATPase that interacts with a network of cofactors and is active in a wide range of ubiquitin-dependent cellular processes. We demonstrate that PV ubiquitination is a pre-requisite for recruitment of these host defense proteins, and their deposition directs Tg PVs to acidification in endothelial cells. We show that p97/VCP universally targets PVs in human cells and restricts Tg in different human cell types. Overall, these findings reveal new players of intracellular host defense of a vacuolated pathogen.


Asunto(s)
Parásitos , Toxoplasma , Animales , Humanos , Toxoplasma/metabolismo , Interferones/metabolismo , Vacuolas/metabolismo , Células Endoteliales , Interferón gamma , Proteína que Contiene Valosina/metabolismo
11.
Science ; 382(6666): eadg2253, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37797010

RESUMEN

Disruption of cellular activities by pathogen virulence factors can trigger innate immune responses. Interferon-γ (IFN-γ)-inducible antimicrobial factors, such as the guanylate binding proteins (GBPs), promote cell-intrinsic defense by attacking intracellular pathogens and by inducing programmed cell death. Working in human macrophages, we discovered that GBP1 expression in the absence of IFN-γ killed the cells and induced Golgi fragmentation. IFN-γ exposure improved macrophage survival through the activity of the kinase PIM1. PIM1 phosphorylated GBP1, leading to its sequestration by 14-3-3σ, which thereby prevented GBP1 membrane association. During Toxoplasma gondii infection, the virulence protein TgIST interfered with IFN-γ signaling and depleted PIM1, thereby increasing GBP1 activity. Although infected cells can restrain pathogens in a GBP1-dependent manner, this mechanism can protect uninfected bystander cells. Thus, PIM1 can provide a bait for pathogen virulence factors, guarding the integrity of IFN-γ signaling.


Asunto(s)
Proteínas de Unión al GTP , Interacciones Huésped-Patógeno , Inmunidad Innata , Interferón gamma , Proteínas Proto-Oncogénicas c-pim-1 , Toxoplasma , Toxoplasmosis , Humanos , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Interferón gamma/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Toxoplasmosis/inmunología , Factores de Virulencia/metabolismo , Macrófagos/inmunología , Proteínas 14-3-3/metabolismo , Interacciones Huésped-Patógeno/inmunología
12.
Nat Commun ; 12(1): 2043, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824312

RESUMEN

The tumour suppressor FBW7 is a substrate adaptor for the E3 ubiquitin ligase complex SKP1-CUL1-F-box (SCF), that targets several oncoproteins for proteasomal degradation. FBW7 is widely mutated and FBW7 protein levels are commonly downregulated in cancer. Here, using an shRNA library screen, we identify the HECT-domain E3 ubiquitin ligase TRIP12 as a negative regulator of FBW7 stability. We find that SCFFBW7-mediated ubiquitylation of FBW7 occurs preferentially on K404 and K412, but is not sufficient for its proteasomal degradation, and in addition requires TRIP12-mediated branched K11-linked ubiquitylation. TRIP12 inactivation causes FBW7 protein accumulation and increased proteasomal degradation of the SCFFBW7 substrate Myeloid Leukemia 1 (MCL1), and sensitizes cancer cells to anti-tubulin chemotherapy. Concomitant FBW7 inactivation rescues the effects of TRIP12 deficiency, confirming FBW7 as an essential mediator of TRIP12 function. This work reveals an unexpected complexity of FBW7 ubiquitylation, and highlights branched ubiquitylation as an important signalling mechanism regulating protein stability.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Biocatálisis , Resistencia a Antineoplásicos , Células HCT116 , Células HEK293 , Humanos , Lisina/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , ARN Interferente Pequeño/metabolismo , Especificidad por Sustrato , Enzimas Ubiquitina-Conjugadoras/metabolismo
13.
BMC Microbiol ; 10: 44, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-20149234

RESUMEN

BACKGROUND: Salmonella enterica serovar Typhimurium (S. Typhimurium) is a major cause of human gastroenteritis worldwide. The outer membrane proteins expressed by S. Typhimurium mediate the process of adhesion and internalisation within the intestinal epithelium of the host thus influencing the progression of disease. Since the outer membrane proteins are surface-exposed, they provide attractive targets for the development of improved antimicrobial agents and vaccines. Various techniques have been developed for their characterisation, but issues such as carryover of cytosolic proteins still remain a problem. In this study we attempted to characterise the surface proteome of S. Typhimurium using Lipid-based Protein Immobilisation technology in the form of LPI FlowCells. No detergents are required and no sample clean up is needed prior to downstream analysis. The immobilised proteins can be digested with proteases in multiple steps to increase sequence coverage, and the peptides eluted can be characterised directly by liquid chromatography - tandem mass spectrometry (LC-MS/MS) and identified from mass spectral database searches. RESULTS: In this study, 54 outer membrane proteins, were identified with two or more peptide hits using a multi-step digest approach. Out of these 28 were lipoproteins, nine were involved in transport and three with enzyme activity These included the transporters BtuB which is responsible for the uptake of vitamin B12, LamB which is involved in the uptake of maltose and maltodextrins and LolB which is involved in the incorporation of lipoproteins in the outer membrane. Other proteins identified included the enzymes MltC which may play a role in cell elongation and division and NlpD which is involved in catabolic processes in cell wall formation as well as proteins involved in virulence such as Lpp1, Lpp2 and OmpX. CONCLUSION: Using a multi-step digest approach the LPI technique enables the incorporation of a multi-step protease work flow ensuring enough sequence coverage of membrane proteins subsequently leading to the identification of more membrane proteins with higher confidence. Compared to current sub-cellular fractionation procedures and previous published work, the LPI technique currently provides the widest coverage of outer membrane proteins identified as demonstrated here for Salmonella Typhimurium.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas Inmovilizadas/química , Salmonella typhi/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Carbonatos/química , Fraccionamiento Celular/métodos , Cromatografía Liquida , Proteínas Inmovilizadas/metabolismo , Lipoproteínas/química , Lipoproteínas/metabolismo , Espectrometría de Masas , Microscopía Electrónica , Proteoma/análisis , Proteómica/métodos , Salmonella typhi/metabolismo , Tensoactivos/química
14.
Cell Rep ; 32(6): 108008, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32783936

RESUMEN

Interferon-inducible guanylate-binding proteins (GBPs) promote cell-intrinsic defense through host cell death. GBPs target pathogens and pathogen-containing vacuoles and promote membrane disruption for release of microbial molecules that activate inflammasomes. GBP1 mediates pyroptosis or atypical apoptosis of Salmonella Typhimurium (STm)- or Toxoplasma gondii (Tg)- infected human macrophages, respectively. The pathogen-proximal detection-mechanisms of GBP1 remain poorly understood, as humans lack functional immunity-related GTPases (IRGs) that assist murine Gbps. Here, we establish that GBP1 promotes the lysis of Tg-containing vacuoles and parasite plasma membranes, releasing Tg-DNA. In contrast, we show GBP1 targets cytosolic STm and recruits caspase-4 to the bacterial surface for its activation by lipopolysaccharide (LPS), but does not contribute to bacterial vacuole escape. Caspase-1 cleaves and inactivates GBP1, and a cleavage-deficient GBP1D192E mutant increases caspase-4-driven pyroptosis due to the absence of feedback inhibition. Our studies elucidate microbe-specific roles of GBP1 in infection detection and its triggering of the assembly of divergent caspase signaling platforms.


Asunto(s)
Caspasas/inmunología , Proteínas de Unión al GTP/inmunología , Salmonella typhimurium/inmunología , Toxoplasma/inmunología , Muerte Celular/inmunología , Células HEK293 , Humanos , Inflamasomas/inmunología , Interferón gamma/farmacología , Ligandos , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Células THP-1 , Toxoplasma/genética , Toxoplasmosis/inmunología , Toxoplasmosis/microbiología , Vacuolas/inmunología
15.
Life Sci Alliance ; 2(6)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31792063

RESUMEN

Determining the exact targets and mechanisms of action of drug molecules that modulate circadian rhythms is critical to develop novel compounds to treat clock-related disorders. Here, we have used phenotypic proteomic profiling (PPP) to systematically determine molecular targets of four circadian period-lengthening compounds in human cells. We demonstrate that the compounds cause similar changes in phosphorylation and activity of several proteins and kinases involved in vital pathways, including MAPK, NGF, B-cell receptor, AMP-activated protein kinases (AMPKs), and mTOR signaling. Kinome profiling further indicated inhibition of CKId, ERK1/2, CDK2/7, TNIK, and MST4 kinases as a common mechanism of action for these clock-modulating compounds. Pharmacological or genetic inhibition of several convergent kinases lengthened circadian period, establishing them as novel circadian targets. Finally, thermal stability profiling revealed binding of the compounds to clock regulatory kinases, signaling molecules, and ubiquitination proteins. Thus, phenotypic proteomic profiling defines novel clock effectors that could directly inform precise therapeutic targeting of the circadian system in humans.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Adenina/análogos & derivados , Adenina/farmacología , Antracenos/farmacología , Línea Celular Tumoral , Relojes Circadianos/efectos de los fármacos , Ritmo Circadiano/genética , Humanos , Fenotipo , Fosforilación , Proteómica , Purinas/farmacología , Roscovitina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/genética
16.
Elife ; 82019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31264961

RESUMEN

Several enzymes can simultaneously interact with multiple intracellular metabolites, however, how the allosteric effects of distinct ligands are integrated to coordinately control enzymatic activity remains poorly understood. We addressed this question using, as a model system, the glycolytic enzyme pyruvate kinase M2 (PKM2). We show that the PKM2 activator fructose 1,6-bisphosphate (FBP) alone promotes tetramerisation and increases PKM2 activity, but addition of the inhibitor L-phenylalanine (Phe) prevents maximal activation of FBP-bound PKM2 tetramers. We developed a method, AlloHubMat, that uses eigenvalue decomposition of mutual information derived from molecular dynamics trajectories to identify residues that mediate FBP-induced allostery. Experimental mutagenesis of these residues identified PKM2 variants in which activation by FBP remains intact but cannot be attenuated by Phe. Our findings reveal residues involved in FBP-induced allostery that enable the integration of allosteric input from Phe and provide a paradigm for the coordinate regulation of enzymatic activity by simultaneous allosteric inputs.


Asunto(s)
Regulación Alostérica , Proteínas Portadoras/metabolismo , Regulación Enzimológica de la Expresión Génica , Proteínas de la Membrana/metabolismo , Hormonas Tiroideas/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular , Análisis Mutacional de ADN , Activadores de Enzimas/metabolismo , Inhibidores Enzimáticos/metabolismo , Fructosadifosfatos/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Simulación de Dinámica Molecular , Fenilalanina/metabolismo , Multimerización de Proteína , Análisis Espectral , Hormonas Tiroideas/química , Hormonas Tiroideas/genética , Proteínas de Unión a Hormona Tiroide
17.
J Exp Med ; 216(2): 450-465, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30642944

RESUMEN

Lung squamous cell carcinoma (LSCC) and adenocarcinoma (LADC) are the most common lung cancer subtypes. Molecular targeted treatments have improved LADC patient survival but are largely ineffective in LSCC. The tumor suppressor FBW7 is commonly mutated or down-regulated in human LSCC, and oncogenic KRasG12D activation combined with Fbxw7 inactivation in mice (KF model) caused both LSCC and LADC. Lineage-tracing experiments showed that CC10+, but not basal, cells are the cells of origin of LSCC in KF mice. KF LSCC tumors recapitulated human LSCC resistance to cisplatin-based chemotherapy, and we identified LUBAC-mediated NF-κB signaling as a determinant of chemotherapy resistance in human and mouse. Inhibition of NF-κB activation using TAK1 or LUBAC inhibitors resensitized LSCC tumors to cisplatin, suggesting a future avenue for LSCC patient treatment.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Resistencia a Antineoplásicos , Neoplasias Pulmonares/enzimología , Complejos Multienzimáticos/metabolismo , Ubiquitinación , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/enzimología , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Cisplatino/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Complejos Multienzimáticos/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
18.
BMC Res Notes ; 11(1): 166, 2018 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-29510761

RESUMEN

OBJECTIVE: The intracellular parasite Toxoplasma gondii can invade any nucleated cell residing inside a parasitophorous vacuole (PV). Upon infection, the cytokine interferon gamma (IFNγ) is produced and elicits host defence mechanisms able to recognise the PV and destroy the parasite. Hereby, Guanylate binding proteins, ubiquitin and the E3 ubiquitin ligases Tripartite Motif Containing 21 (TRIM21) and TNF receptor associated factor 6 are targeted to the murine PV leading to its destruction. This study is the side product of research aiming to identify ubiquitinated substrates in a TRIM21-dependent fashion in murine cells infected with Toxoplasma. RESULTS: We infected IFNγ-stimulated murine embryonic fibroblasts (MEFs) from either C57BL/6×129 wild-type (WT) mice or C57BL/6 TRIM21-/- mice with Toxoplasma. Using mass spectrometry, we analysed proteins in both cell backgrounds presenting with the di-glycine remnant of ubiquitination. In addition, we compared peptide levels between WT and TRIM21-/- cells. In line with earlier reports, Gbp1 was expressed to higher levels in the C57BL/6×129 WT MEFs compared to the C57BL/6-only background TRIM21-/- MEFs. Protein expression differences in these different murine backgrounds thus precluded identification of TRIM21-dependent ubiquitinated substrates. Nevertheless, we identified and confirmed Gbp1 and Gbp2 as being ubiquitinated in a Toxoplasma-infection independent manner.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Toxoplasmosis , Ubiquitinación , Animales , Embrión de Mamíferos , Fibroblastos , Ratones , Ratones Endogámicos C57BL
19.
Dev Cell ; 47(5): 645-659.e6, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30393076

RESUMEN

Meiotic synapsis and recombination ensure correct homologous segregation and genetic diversity. Asynapsed homologs are transcriptionally inactivated by meiotic silencing, which serves a surveillance function and in males drives meiotic sex chromosome inactivation. Silencing depends on the DNA damage response (DDR) network, but how DDR proteins engage repressive chromatin marks is unknown. We identify the histone H3-lysine-9 methyltransferase SETDB1 as the bridge linking the DDR to silencing in male mice. At the onset of silencing, X chromosome H3K9 trimethylation (H3K9me3) enrichment is downstream of DDR factors. Without Setdb1, the X chromosome accrues DDR proteins but not H3K9me3. Consequently, sex chromosome remodeling and silencing fail, causing germ cell apoptosis. Our data implicate TRIM28 in linking the DDR to SETDB1 and uncover additional factors with putative meiotic XY-silencing functions. Furthermore, we show that SETDB1 imposes timely expression of meiotic and post-meiotic genes. Setdb1 thus unites the DDR network, asynapsis, and meiotic chromosome silencing.


Asunto(s)
Emparejamiento Cromosómico , Daño del ADN , Silenciador del Gen , Código de Histonas , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Apoptosis , Reparación del ADN , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína 28 que Contiene Motivos Tripartito/genética , Proteína 28 que Contiene Motivos Tripartito/metabolismo
20.
Cell Rep ; 18(10): 2480-2493, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28273461

RESUMEN

Cellular senescence is an important in vivo mechanism that prevents the propagation of damaged cells. However, the precise mechanisms regulating senescence are not well characterized. Here, we find that ITGB3 (integrin beta 3 or ß3) is regulated by the Polycomb protein CBX7. ß3 expression accelerates the onset of senescence in human primary fibroblasts by activating the transforming growth factor ß (TGF-ß) pathway in a cell-autonomous and non-cell-autonomous manner. ß3 levels are dynamically increased during oncogene-induced senescence (OIS) through CBX7 Polycomb regulation, and downregulation of ß3 levels overrides OIS and therapy-induced senescence (TIS), independently of its ligand-binding activity. Moreover, cilengitide, an αvß3 antagonist, has the ability to block the senescence-associated secretory phenotype (SASP) without affecting proliferation. Finally, we show an increase in ß3 levels in a subset of tissues during aging. Altogether, our data show that integrin ß3 subunit is a marker and regulator of senescence.


Asunto(s)
Senescencia Celular , Integrina beta3/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Anciano de 80 o más Años , Envejecimiento/metabolismo , Animales , Línea Celular , Células Cultivadas , Senescencia Celular/genética , Niño , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Integrina beta3/genética , Marcaje Isotópico , Ratones , Complejo Represivo Polycomb 1/metabolismo , Subunidades de Proteína/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA