Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.153
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 38(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30944096

RESUMEN

Astrocytes are critical regulators of neuroinflammation in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Growing evidence indicates that ubiquitination of signaling molecules is an important cell-intrinsic mechanism governing astrocyte function during MS and EAE Here, we identified an upregulation of the deubiquitinase OTU domain, ubiquitin aldehyde binding 1 (OTUB1) in astrocytes during MS and EAE Mice with astrocyte-specific OTUB1 ablation developed more severe EAE due to increased leukocyte accumulation, proinflammatory gene transcription, and demyelination in the spinal cord as compared to control mice. OTUB1-deficient astrocytes were hyperactivated in response to IFN-γ, a fingerprint cytokine of encephalitogenic T cells, and produced more proinflammatory cytokines and chemokines than control astrocytes. Mechanistically, OTUB1 inhibited IFN-γ-induced Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling by K48 deubiquitination and stabilization of the JAK2 inhibitor suppressor of cytokine signaling 1 (SOCS1). Thus, astrocyte-specific OTUB1 is a critical inhibitor of neuroinflammation in CNS autoimmunity.


Asunto(s)
Astrocitos/inmunología , Astrocitos/patología , Autoinmunidad/genética , Cisteína Endopeptidasas/fisiología , Interferón gamma/fisiología , Inflamación Neurogénica/genética , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Células Cultivadas , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Interferón gamma/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inflamación Neurogénica/patología , Neuroinmunomodulación/genética
2.
J Virol ; 95(14): e0015021, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33952634

RESUMEN

Potyviridae is the largest family of plant RNA viruses. Their genomes are expressed through long polyproteins that are usually headed by the leader endopeptidase P1. This protein can be classified as type A or type B based on host proteolytic requirements and RNA silencing suppression (RSS) capacity. The main Potyviridae genus is Potyvirus, and a group of potyviruses infecting sweet potato presents an enlarged P1 protein with a polymerase slippage motif that produces an extra product termed P1N-PISPO. These two proteins display some RSS activity and are expressed followed by HCPro, which appears to be the main RNA silencing suppressor in these viruses. Here, we studied the behavior of the P1 protein of Sweet potato feathery mottle virus (SPFMV) using a viral system based on a canonical potyvirus, Plum pox virus (PPV), and discovered that this protein is able to replace both PPV P1 and HCPro. We also found that P1N-PISPO, produced after polymerase slippage, provides extra RNA silencing suppression capacity to SPFMV P1 in this viral context. In addition, the results showed that presence of two type A P1 proteins was detrimental for viral viability. The ample recombination spectrum that we found in the recovered viruses supports the strong adaptation capacity of P1 proteins and signals the N-terminal part of SPFMV P1 as essential for RSS activity. Further analyses provided data to add extra layers to the evolutionary history of sweet potato-infecting potyvirids. IMPORTANCE Plant viruses represent a major challenge for agriculture worldwide and Potyviridae, being the largest family of plant RNA viruses, is one of the primary players. P1, the leader endopeptidase, is a multifunctional protein that contributes to the successful spread of these viruses over a wide host range. Understanding how P1 proteins work, their dynamic interplay during viral infection, and their evolutionary path is critical for the development of strategic tools to fight the multiple diseases these viruses cause. We focused our efforts on the P1 protein of Sweet potato feathery mottle virus, which is coresponsible for the most devastating disease in sweet potato. The significance of our research is in understanding the capacity of this protein to perform several independent functions, using this knowledge to learn more about P1 proteins in general and the potyvirids infecting this host.


Asunto(s)
Adaptación Fisiológica , Cisteína Endopeptidasas/genética , Ipomoea batatas/virología , Virus Eruptivo de la Ciruela/fisiología , Potyvirus/fisiología , Proteínas Virales/genética , Cisteína Endopeptidasas/fisiología , Prueba de Complementación Genética , Enfermedades de las Plantas/virología , Plásmidos , Virus Eruptivo de la Ciruela/genética , Potyvirus/genética , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Virus Reordenados/genética , Virus Reordenados/fisiología , Proteínas Virales/fisiología
3.
FASEB J ; 35(12): e22039, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34793600

RESUMEN

OTUB1 is one of the most highly expressed deubiquitinases, counter-regulating the two most abundant ubiquitin chain types. OTUB1 expression is linked to the development and progression of lung cancer and idiopathic pulmonary fibrosis in humans. However, the physiological function of OTUB1 is unknown. Here, we show that constitutive whole-body Otub1 deletion in mice leads to perinatal lethality by asphyxiation. Analysis of (single-cell) RNA sequencing and proteome data demonstrated that OTUB1 is expressed in all lung cell types with a particularly high expression during late-stage lung development (E16.5, E18.5). At E18.5, the lungs of animals with Otub1 deletion presented with increased cell proliferation that decreased saccular air space and prevented inhalation. Flow cytometry-based analysis of E18.5 lung tissue revealed that Otub1 deletion increased proliferation of major lung parenchymal and mesenchymal/other non-hematopoietic cell types. Adult mice with conditional whole-body Otub1 deletion (wbOtub1del/del ) also displayed increased lung cell proliferation in addition to hyperventilation and failure to adapt the respiratory pattern to hypoxia. On the molecular level, Otub1 deletion enhanced mTOR signaling in embryonic and adult lung tissues. Based on these results, we propose that OTUB1 is a negative regulator of mTOR signaling with essential functions for lung cell proliferation, lung development, adult lung tissue homeostasis, and respiratory regulation.


Asunto(s)
Proliferación Celular , Cisteína Endopeptidasas/fisiología , Homeostasis , Hiperventilación/patología , Enfermedades Pulmonares/patología , Insuficiencia Respiratoria/patología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Femenino , Hiperventilación/etiología , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Insuficiencia Respiratoria/etiología , Serina-Treonina Quinasas TOR/genética
4.
Mol Cell ; 55(1): 47-58, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24930734

RESUMEN

The ubiquitin-like SUMO system regulates gene expression, but the molecular insights into this process are incomplete. We show that the SUMO-specific isopeptidase SENP3 controls H3K4 methylation by regulating histone-modifying SET1/MLL complexes. SET1/MLL complexes are composed of a histone methyltransferase and the regulatory components WDR5, RbBP5, Ash2L, and DPY-30. MLL1/MLL2 complexes contain menin as additional component and are particularly important for the activation of HOX genes. We demonstrate that SENP3 is associated with MLL1/MLL2 complexes and catalyzes deSUMOylation of RbBP5. This is required for activation of a subset of HOX genes, including the developmental regulator DLX3. In the absence of SENP3, the association of menin and Ash2L with the DLX3 gene is impaired, leading to decreased H3K4 methylation and reduced recruitment of active RNA polymerase II. Importantly, the SENP3-DLX3 pathway dictates osteogenic differentiation of human stem cells, thus delineating the importance of balanced SUMOylation for epigenetic control of gene expression programs.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Neoplasias/metabolismo , Osteogénesis/genética , Diferenciación Celular/genética , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al ADN/genética , Saco Dental/citología , Saco Dental/metabolismo , Células HeLa , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio/genética , Humanos , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas , Células Madre/citología , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Biochem Biophys Res Commun ; 532(4): 640-646, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32912629

RESUMEN

Endothelial cells injury and pro-inflammation cytokines release are the initial steps of hyperhomocysteinemia (HHcy)-associated vascular inflammation. Pyroptosis is a newly identified pro-inflammation form of programmed cell death, causing cell lysis and IL-1ß release, and characterized by the caspases-induced cleavage of its effector molecule gasdermins (GSDMs). However, the effect of homocysteine (Hcy) on endothelial cells pyroptosis and the underlying mechanisms have not been fully defined. We have previously reported that Hcy induces vascular endothelial inflammation accompanied by the increase of high mobility group box-1 protein (HMGB1) and lysosomal cysteine protease cathepsin V in endothelial cells, and other studies have shown that HMGB1 or cathepsins are involved in activation of NLRP3 inflammasome and caspase-1. Here, we investigated the role of HMGB1 and cathepsin V in the process of Hcy-induced pyroptosis. We observed an increase in plasma IL-1ß levels in HHcy patients and mice models, cathepsin V inhibitor reduced the plasma IL-1ß levels and cleavage of GSDMD full-length into GSDMD N-terminal in the thoracic aorta of hyperhomocysteinemia mice. Using cultured HUVECs, we observed that Hcy promoted GSDMD N-terminal expression, silencing GSDMD or HMGB1 rescued Hcy-induced pyroptosis. HMGB1 also increased GSDMD N-terminal expression, and silencing cathepsin V reversed HMGB1-induced pyroptosis. HMGB1 could increase lysosome permeability, and silencing cathepsin V attenuated HMGB1-induced activation of caspase-1. In conclusion, this study has delineated a novel mechanism that HMGB1 mediated Hcy-induced endothelial cells pyroptosis partly via cathepsin V-dependent pathway.


Asunto(s)
Catepsinas/fisiología , Cisteína Endopeptidasas/fisiología , Endotelio Vascular/citología , Proteína HMGB1/fisiología , Homocisteína/fisiología , Piroptosis , Anciano , Animales , Caspasa 1/metabolismo , Línea Celular , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/metabolismo , Interleucina-1beta/sangre , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas de Unión a Fosfato/metabolismo , Arterias Torácicas/metabolismo
6.
PLoS Pathog ; 14(2): e1006901, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29415027

RESUMEN

Stress granules (SGs) contain stalled messenger ribonucleoprotein complexes and are related to the regulation of mRNA translation. Picornavirus infection can interfere with the formation of SGs. However, the detailed molecular mechanisms and functions of picornavirus-mediated regulation of SG formation are not clear. Here, we found that the 2A protease of a picornavirus, EV71, induced atypical stress granule (aSG), but not typical stress granule (tSG), formation via cleavage of eIF4GI. Furthermore, 2A was required and sufficient to inhibit tSGs induced by EV71 infection, sodium arsenite, or heat shock. Infection of 2A protease activity-inactivated recombinant EV71 (EV71-2AC110S) failed to induce aSG formation and only induced tSG formation, which is PKR and eIF2α phosphorylation-dependent. By using a Renilla luciferase mRNA reporter system and RNA fluorescence in situ hybridization assay, we found that EV71-induced aSGs were beneficial to viral translation through sequestering only cellular mRNAs, but not viral mRNAs. In addition, we found that the 2A protease of other picornaviruses such as poliovirus and coxsackievirus also induced aSG formation and blocked tSG formation. Taken together, our results demonstrate that, on one hand, EV71 infection induces tSG formation via the PKR-eIF2α pathway, and on the other hand, 2A, but not 3C, blocks tSG formation. Instead, 2A induces aSG formation by cleaving eIF4GI to sequester cellular mRNA but release viral mRNA, thereby facilitating viral translation.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Gránulos Citoplasmáticos/metabolismo , Interacciones Huésped-Patógeno , Picornaviridae/enzimología , Estrés Fisiológico/fisiología , Proteínas Virales/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Células HEK293 , Células HeLa , Humanos , Picornaviridae/metabolismo , Biosíntesis de Proteínas , Proteolisis
7.
PLoS Pathog ; 14(9): e1007296, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30248143

RESUMEN

SARS-coronavirus (CoV) is a zoonotic agent derived from rhinolophid bats, in which a plethora of SARS-related, conspecific viral lineages exist. Whereas the variability of virulence among reservoir-borne viruses is unknown, it is generally assumed that the emergence of epidemic viruses from animal reservoirs requires human adaptation. To understand the influence of a viral factor in relation to interspecies spillover, we studied the papain-like protease (PLP) of SARS-CoV. This key enzyme drives the early stages of infection as it cleaves the viral polyprotein, deubiquitinates viral and cellular proteins, and antagonizes the interferon (IFN) response. We identified a bat SARS-CoV PLP, which shared 86% amino acid identity with SARS-CoV PLP, and used reverse genetics to insert it into the SARS-CoV genome. The resulting virus replicated like SARS-CoV in Vero cells but was suppressed in IFN competent MA-104 (3.7-fold), Calu-3 (2.6-fold) and human airway epithelial cells (10.3-fold). Using ectopically-expressed PLP variants as well as full SARS-CoV infectious clones chimerized for PLP, we found that a protease-independent, anti-IFN function exists in SARS-CoV, but not in a SARS-related, bat-borne virus. This PLP-mediated anti-IFN difference was seen in primate, human as well as bat cells, thus independent of the host context. The results of this study revealed that coronavirus PLP confers a variable virulence trait among members of the species SARS-CoV, and that a SARS-CoV lineage with virulent PLPs may have pre-existed in the reservoir before onset of the epidemic.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Proteínas Virales/fisiología , Secuencia de Aminoácidos , Animales , Quirópteros/virología , Chlorocebus aethiops , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/genética , Reservorios de Enfermedades/virología , Células HEK293 , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Interferones/antagonistas & inhibidores , Filogenia , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Homología de Secuencia de Aminoácido , Síndrome Respiratorio Agudo Grave/epidemiología , Síndrome Respiratorio Agudo Grave/virología , Ubiquitina/metabolismo , Células Vero , Proteínas Virales/genética , Virulencia/genética , Virulencia/fisiología , Replicación Viral/genética , Replicación Viral/fisiología , Zoonosis/epidemiología , Zoonosis/virología
8.
Arterioscler Thromb Vasc Biol ; 39(4): e130-e145, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30676070

RESUMEN

Objective- Macrophages participate in the pathogenesis of pulmonary arterial hypertension (PAH). Lgmn (Legumain), a newly discovered cysteine proteinase belonging to the C13 peptidase family, is primarily expressed in macrophages; however, its roles in PAH remain unknown. Approach and Results- Herein, Lgmn was upregulated in lung tissues of PAH mice subjected to hypoxia plus SU5416 and PAH rats challenged with monocrotaline. Global Lgmn ablation and macrophage-specific ablation alleviated PAH compared with wild-type mice, evident from a reduction in right ventricular systolic pressure, the ratio of the right ventricular wall to the left ventricular wall plus the septum, the pulmonary vascular media thickness, and pulmonary vascular muscularization. Increased expression of ECM (extracellular matrix) proteins was correlated with MMP (matrix metalloproteinase)-2 activation and TGF (transforming growth factor)-ß1 signaling in the PAs. Although Lgmn did not affect inflammatory cell infiltration and PA smooth muscle cell proliferation, it drove increased the synthesis of ECM proteins via MMP-2 activation. MMP-2 hydrolyzed the TGF-ß1 precursor to the active form. An Lgmn-specific inhibitor markedly ameliorated PAH. Clinically, serum Lgmn levels were closely associated with the severity of idiopathic PAH. Conclusions- Our results indicate that Lgmn inhibition could be an effective strategy for preventing or delaying PAH.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Hipertensión Pulmonar/enzimología , Macrófagos/enzimología , Metaloproteinasa 2 de la Matriz/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Animales , Inhibidores de Caspasas/farmacología , Cisteína Endopeptidasas/deficiencia , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/prevención & control , Hipoxia/enzimología , Indoles/toxicidad , Inflamación , Pulmón/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Monocrotalina/toxicidad , Pirroles/toxicidad , Ratas , Índice de Severidad de la Enfermedad , Transducción de Señal , Remodelación Vascular/fisiología
9.
Infect Immun ; 87(5)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30804098

RESUMEN

The human gastrointestinal tract (GIT) is inhabited by a dense microbial community of symbionts. Enterococci are among the earliest members of this community and remain core members of the GIT microbiota throughout life. Enterococci have also recently emerged as opportunistic pathogens and major causes of nosocomial infections. Although recognized as a prerequisite for infection, colonization of the GIT by enterococci remains poorly understood. One way that bacteria adapt to dynamic ecosystems like the GIT is through the use of their surface proteins to sense and interact with components of their immediate environment. In Gram-positive bacteria, a subset of surface proteins relies on an enzyme called sortase for covalent attachment to the cell wall. Here, we show that the housekeeping sortase A (SrtA) enzyme promotes intestinal colonization by enterococci. Furthermore, we show that the enzymatic activity of SrtA is key to the ability of Enterococcus faecalis to bind mucin (a major component of the GIT mucus). We also report the GIT colonization phenotypes of E. faecalis mutants lacking selected sortase-dependent proteins (SDPs). Further examination of the mucin binding ability of these mutants suggests that adhesion to mucin contributes to intestinal colonization by E. faecalis.


Asunto(s)
Aminoaciltransferasas/fisiología , Proteínas Bacterianas/fisiología , Pared Celular/efectos de los fármacos , Cisteína Endopeptidasas/fisiología , Enterococcus/fisiología , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/fisiología , Animales , Modelos Animales de Enfermedad , Tracto Gastrointestinal/microbiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
10.
Mol Cell ; 43(1): 57-71, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21726810

RESUMEN

Caspase-2 is an evolutionarily conserved caspase, yet its biological function and cleavage targets are poorly understood. Caspase-2 is activated by the p53 target gene product PIDD (also known as LRDD) in a complex called the Caspase-2-PIDDosome. We show that PIDD expression promotes growth arrest and chemotherapy resistance by a mechanism that depends on Caspase-2 and wild-type p53. PIDD-induced Caspase-2 directly cleaves the E3 ubiquitin ligase Mdm2 at Asp 367, leading to loss of the C-terminal RING domain responsible for p53 ubiquitination. As a consequence, N-terminally truncated Mdm2 binds p53 and promotes its stability. Upon DNA damage, p53 induction of the Caspase-2-PIDDosome creates a positive feedback loop that inhibits Mdm2 and reinforces p53 stability and activity, contributing to cell survival and drug resistance. These data establish Mdm2 as a cleavage target of Caspase-2 and provide insight into a mechanism of Mdm2 inhibition that impacts p53 dynamics upon genotoxic stress.


Asunto(s)
Caspasa 2/fisiología , Cisteína Endopeptidasas/fisiología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Caspasa 2/metabolismo , Cisplatino/farmacología , Cisteína Endopeptidasas/metabolismo , Daño del ADN , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Retroalimentación Fisiológica , Humanos
11.
Nucleic Acids Res ; 45(11): 6729-6745, 2017 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-28379520

RESUMEN

Pre-mRNA splicing is catalyzed by the spliceosome, a multi-megadalton ribonucleoprotein machine. Previous work from our laboratory revealed the splicing factor SRSF1 as a regulator of the SUMO pathway, leading us to explore a connection between this pathway and the splicing machinery. We show here that addition of a recombinant SUMO-protease decreases the efficiency of pre-mRNA splicing in vitro. By mass spectrometry analysis of anti-SUMO immunoprecipitated proteins obtained from purified splicing complexes formed along the splicing reaction, we identified spliceosome-associated SUMO substrates. After corroborating SUMOylation of Prp3 in cultured cells, we defined Lys 289 and Lys 559 as bona fide SUMO attachment sites within this spliceosomal protein. We further demonstrated that a Prp3 SUMOylation-deficient mutant while still capable of interacting with U4/U6 snRNP components, is unable to co-precipitate U2 and U5 snRNA and the spliceosomal proteins U2-SF3a120 and U5-Snu114. This SUMOylation-deficient mutant fails to restore the splicing of different pre-mRNAs to the levels achieved by the wild type protein, when transfected into Prp3-depleted cultured cells. This mutant also shows a diminished recruitment to active spliceosomes, compared to the wild type protein. These findings indicate that SUMO conjugation plays a role during the splicing process and suggest the involvement of Prp3 SUMOylation in U4/U6•U5 tri-snRNP formation and/or recruitment.


Asunto(s)
Proteínas Nucleares/metabolismo , Empalme del ARN , Ribonucleoproteína Nuclear Pequeña U4-U6/metabolismo , Empalmosomas/metabolismo , Sumoilación , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/fisiología , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/química , Precursores del ARN/química , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Ribonucleoproteína Nuclear Pequeña U4-U6/química
12.
Rev Esp Enferm Dig ; 111(3): 214-222, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30507245

RESUMEN

BACKGROUND AND AIMS: pathological angiogenesis plays an important role in the progression of chronic liver diseases. Asparaginyl endopeptidase (AEP) participates in tumor angiogenesis and was recently shown to be associated with liver fibrosis. This study aimed to explore the effect of AEP on liver sinusoidal endothelial cell (LSECs) angiogenesis and determine the underlying mechanism. METHODS: cultured LSECs were infected with lentiviruses in order to suppress AEP expression (AEP-KD1, AEP-KD2). The effect of AEP on LSECs proliferation, apoptosis and migration were subsequently determined by a CCK8 assay, flow cytometry and wound-healing and Transwell assays, respectively, in AEP knocked-down and control LSECs. The expression of the endothelial cell surface markers CD31, CD34 and von Willebrand factor (vWF) were detected by immunofluorescence assay and western blot. The angiogenic factors, vascular endothelial growth factor receptor 2 (VEGFR2) and interleukin 8 (IL 8) were detected by real-time PCR and western blot. The effect of AEP on vessel tube formation by LSECs was examined by Matrigel™ tube-formation assay. Phosphoinositide 3-kinase (PI3K)/Akt expression and phosphorylation were detected by western blot. RESULTS: AEP was effectively knocked down by lentivirus infection in LSECs. Down-regulation of AEP expression significantly decreased proliferation and migration and increased apoptosis of LSECs. Moreover, expression levels of the endothelial cell surface markers CD31, CD34 and vWF, as well as angiogenic factors VEGFR2 and IL 8, were also reduced after AEP was knocked-down. The vessel tube formation abilities of AEP-KD1 and AEP-KD2 LSECs were significantly inhibited compared with LSECs without AEP knocked-down. Down-regulation of AEP also inhibited the phosphorylation of PI3K and Akt. CONCLUSION: AEP promotes LSECs angiogenesis in vitro, possibly via the PI3K/Akt pathway. AEP may therefore be a potential therapeutic target for preventing the progression of liver fibrosis.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Hepatocitos/fisiología , Neovascularización Patológica/etiología , Fosfatidilinositol 3-Quinasa/metabolismo , Antígenos CD34/metabolismo , Apoptosis , Movimiento Celular , Proliferación Celular , Células Cultivadas , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/farmacología , Progresión de la Enfermedad , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Interleucina-8/metabolismo , Lentivirus , Neovascularización Patológica/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas , Factor de von Willebrand/metabolismo
13.
Biochemistry ; 57(22): 3176-3190, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29336553

RESUMEN

Cruzain, an important drug target for Chagas disease, is a member of clan CA of the cysteine proteases. Understanding the catalytic mechanism of cruzain is vital to the design of new inhibitors. To this end, we have determined pH-rate profiles for substrates and affinity agents and solvent kinetic isotope effects in pre-steady-state and steady-state modes using three substrates: Cbz-Phe-Arg-AMC, Cbz-Arg-Arg-AMC, and Cbz-Arg-Ala-AMC. The pH-rate profile of kcat/ Km for Cbz-Arg-Arg-AMC indicated p K1 = 6.6 (unprotonated) and p K2 ∼ 9.6 (protonated) groups were required for catalysis. The temperature dependence of the p K = 6.2-6.6 group exhibited a Δ Hion value of 8.4 kcal/mol, typical of histidine. The pH-rate profile of inactivation by iodoacetamide confirmed that the catalytic cysteine possesses a p Ka of 9.8. Normal solvent kinetic isotope effects were observed for both D2O kcat (1.6-2.1) and D2O kcat/ Km (1.1-1.4) for all three substrates. Pre-steady-state kinetics revealed exponential bursts of AMC production for Cbz-Phe-Arg-AMC and Cbz-Arg-Arg-AMC, but not for Cbz-Arg-Ala-AMC. The overall solvent isotope effect on kcat can be attributed to the solvent isotope effect on the deacylation step. Our results suggest that cruzain is unique among papain-like cysteine proteases in that the catalytic cysteine and histidine have neutral charges in the free enzyme. The generation of the active thiolate of the catalytic cysteine is likely preceded (and possibly triggered) by a ligand-induced conformational change, which could bring the catalytic dyad into the proximity to effect proton transfer.


Asunto(s)
Cisteína Endopeptidasas/química , Proteínas Protozoarias/química , Trypanosoma cruzi/enzimología , Caspasas , Catálisis , Cisteína/metabolismo , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/fisiología , Histidina/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Protones , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/fisiología , Solventes , Especificidad por Sustrato , Trypanosoma cruzi/metabolismo
14.
Curr Top Microbiol Immunol ; 404: 159-175, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27097813

RESUMEN

In Gram-positive bacteria, protein precursors with a signal peptide and a cell wall sorting signal (CWSS)-which begins with an LPXTG motif, followed by a hydrophobic domain and a tail of positively charged residues-are targeted to the cell envelope by a transpeptidase enzyme call sortase. Evolution and selective pressure gave rise to six classes of sortase, i.e., SrtA-F. Only class C sortases are capable of polymerizing substrates harboring the pilin motif and CWSS into protein polymers known as pili or fimbriae, whereas the others perform cell wall anchoring functions. Regardless of the products generated from these sortases, the basic principle of sortase-catalyzed transpeptidation is the same. It begins with the cleavage of the LPXTG motif, followed by the cross-linking of this cleaved product at the threonine residue to a nucleophile, i.e., an active amino group of the peptidoglycan stem peptide or the lysine residue of the pilin motif. This chapter will summarize the efforts to identify and characterize sortases and their associated pathways with emphasis on the cell wall anchoring function.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Bacterias Grampositivas/metabolismo , Señales de Clasificación de Proteína/fisiología , Secuencias de Aminoácidos , Aminoaciltransferasas/fisiología , Proteínas Bacterianas/fisiología , Cisteína Endopeptidasas/fisiología
15.
Immunity ; 31(5): 696-8, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19932067

RESUMEN

Toll-like receptor 9 (TLR9) requires proteolytic maturation to acquire signaling capacity; however, the involved protease(s) is unclear. In this issue of Immunity, Sepulveda et al. (2009) demonstrate that in dendritic cells, asparaginyl endopeptidase is a key protease that controls TLR9 maturation.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Células Dendríticas/inmunología , Receptor Toll-Like 9/metabolismo , Humanos , Macrófagos/inmunología , Transducción de Señal
16.
Immunity ; 31(5): 737-48, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19879164

RESUMEN

Intracellular Toll-like receptor 3 (TLR3), TLR7, and TLR9 localize in endosomes and recognize single-stranded RNA and nucleotides from viruses and bacteria. This interaction induces their conformational changes resulting in the production of proinflammatory cytokines and upregulation of cell surface molecules. TLR9 requires a proteolytic cleavage for its signaling. Here, we report that myeloid and plasmacytoid dendritic cells (DCs) deficient for the asparagine endopeptidase (AEP), a cysteine lysosomal protease, showed a decrease in the secretion of proinflammatory cytokines in response to TLR9 stimulation in vitro and in vivo. Upon stimulation, full-length TLR9 was cleaved into a 72 kDa fragment and this processing was strongly reduced in DCs lacking AEP. Processed TLR9 coeluted with the adaptor molecule MyD88 and AEP after size exclusion chromatography. When expressed in AEP-deficient DCs, the 72 kDa proteolytic fragment restored TLR9 signaling. Thus, our results identify an endocytic protease playing a critical role in TLR processing and signaling in DCs.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Células Dendríticas/inmunología , Transducción de Señal , Receptor Toll-Like 9/metabolismo , Animales , Catepsinas/metabolismo , Cisteína Endopeptidasas/genética , Ratones , Ratones Noqueados
17.
Biochim Biophys Acta Gen Subj ; 1862(3): 557-566, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29203282

RESUMEN

BACKGROUND: The development of approaches that increase therapeutic effects of anti-cancer drugs is one of the most important tasks of oncology. Caloric restriction in vivo or serum deprivation (SD) in vitro has been shown to be an effective tool for sensitizing cancer cells to chemotherapeutic drugs. However, the detailed mechanisms underlying the enhancement of apoptosis in cancer cells by SD remain to be elucidated. METHODS: Flow cytometry, caspase activity assay and western blotting were used for cell death rate evaluation. Western blotting, gel-filtration, siRNA approach and qRT-PCR were used to elucidate the mechanism underlying cell death potentiation upon SD. RESULTS: We demonstrated that SD sensitizes cancer cells to treatment with chemotherapeutic agent cisplatin. This effect is independent on activation of caspases-2 and -8, apical caspases triggering apoptosis in response to genotoxic stress. SD potentiates cell death via downregulation of the anti-apoptotic protein Mcl-1. In fact, SD reduces the Mcl-1 mRNA level, which consequently decreases the Mcl-1 protein level and renders cells more susceptible to apoptosis induction via the formation of apoptosome. CONCLUSIONS: Mcl-1 protein is an important regulator of sensitivity of cancer cells to apoptotic stimuli upon SD. GENERAL SIGNIFICANCE: This study identifies Mcl-1 as a new target for the sensitization of human cancer cells to cell death by SD, which is of great significance for the development of efficient anti-cancer therapies.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Medio de Cultivo Libre de Suero/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/biosíntesis , Proteínas de Neoplasias/biosíntesis , Apoptosis/fisiología , Apoptosomas/fisiología , Caspasa 2/fisiología , Caspasa 8/fisiología , Línea Celular Tumoral , Cisteína Endopeptidasas/fisiología , Regulación hacia Abajo , Resistencia a Antineoplásicos/fisiología , Células HeLa , Humanos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/fisiología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética
18.
Proc Natl Acad Sci U S A ; 112(19): 6146-51, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25902531

RESUMEN

At their margins, tumors often contain neutrophils, dendritic cells, and activated macrophages, which express class II MHC and CD11b products. The interplay between stromal cells, tumor cells, and migratory cells such as lymphocytes creates opportunities for noninvasive imaging of immune responses. We developed alpaca-derived antibody fragments specific for mouse class II MHC and CD11b products, expressed on the surface of a variety of myeloid cells. We validated these reagents by flow cytometry and two-photon microscopy to obtain images at cellular resolution. To enable noninvasive imaging of the targeted cell populations, we developed a method to site-specifically label VHHs [the variable domain (VH) of a camelid heavy-chain only antibody] with (18)F or (64)Cu. Radiolabeled VHHs rapidly cleared the circulation (t1/2 ≈ 20 min) and clearly visualized lymphoid organs. We used VHHs to explore the possibility of imaging inflammation in both xenogeneic and syngeneic tumor models, which resulted in detection of tumors with remarkable specificity. We also imaged the infiltration of myeloid cells upon injection of complete Freund's adjuvant. Both anti-class II MHC and anti-CD11b VHHs detected inflammation with excellent specificity. Given the ease of manufacture and labeling of VHHs, we believe that this method could transform the manner in which antitumor responses and/or infectious events may be tracked.


Asunto(s)
Sistema Inmunológico/fisiología , Neoplasias/inmunología , Tomografía de Emisión de Positrones , Aminoaciltransferasas/fisiología , Animales , Anticuerpos/inmunología , Antineoplásicos/uso terapéutico , Proteínas Bacterianas/fisiología , Células de la Médula Ósea/metabolismo , Radioisótopos de Cobre/química , Cisteína Endopeptidasas/fisiología , Citometría de Flujo , Radioisótopos de Flúor/química , Adyuvante de Freund , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Inflamación , Ratones , Ratones Endogámicos C57BL , Células Mieloides/patología , Trasplante de Neoplasias , Neoplasias/terapia
19.
Eur J Immunol ; 45(12): 3257-68, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26399368

RESUMEN

The immunodominant MART-1(26(27)-35) epitope, liberated from the differentiation antigen melanoma antigen recognized by T cells/melanoma antigen A (MART-1/Melan-A), has been frequently targeted in melanoma immunotherapy, but with limited clinical success. Previous studies suggested that this is in part due to an insufficient peptide supply and epitope presentation, since proteasomes containing the immunosubunits ß5i/LMP7 (LMP, low molecular weight protein) or ß1i/LMP2 and ß5i/LMP7 interfere with MART-1(26-35) epitope generation in tumor cells. Here, we demonstrate that in addition the IFN-γ-inducible proteasome subunit ß2i/MECL-1 (multicatalytic endopeptidase complex-like 1), proteasome activator 28 (PA28), and ER-resident aminopeptidase 1 (ERAP1) impair MART-1(26-35) epitope generation. ß2i/MECL-1 and PA28 negatively affect C- and N-terminal cleavage and therefore epitope liberation from the proteasome, whereas ERAP1 destroys the MART-1(26-35) epitope by overtrimming activity. Constitutive expression of PA28 and ERAP1 in melanoma cells indicate that both interfere with MART-1(26-35) epitope generation even in the absence of IFN-γ. In summary, our results provide first evidence that activities of different antigen-processing components contribute to an inefficient MART-1(26-35) epitope presentation, suggesting the tumor cell's proteolytic machinery might have an important impact on the outcome of epitope-specific immunotherapies.


Asunto(s)
Aminopeptidasas/fisiología , Epítopos/inmunología , Melanoma/inmunología , Proteínas Musculares/fisiología , Proteínas de Neoplasias/inmunología , Complejo de la Endopetidasa Proteasomal/fisiología , Linfocitos T/inmunología , Línea Celular Tumoral , Cisteína Endopeptidasas/fisiología , Humanos , Antígenos de Histocompatibilidad Menor
20.
Br J Dermatol ; 174(5): 1030-41, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26713607

RESUMEN

BACKGROUND: Dermatomyositis (DM) and systemic lupus erythematosus (SLE) have common skin features, including dermal mucin deposition and interferon signature, although their roles are unknown. OBJECTIVES: To identify common or specific molecular changes in DM and SLE skin. METHODS: Proteomic analysis was performed using DM and healthy skin. Glycosaminoglycans were analysed by high-performance liquid chromatography. RESULTS: The expression of 60 proteins was upregulated or downregulated in DM skin compared with healthy skin in the proteomic analysis. Among those proteins, PSMB9, an immunoproteasome subunit, was upregulated in the epidermis of DM and SLE, but not in other skin diseases. Furthermore, versican V1, a core protein for glycosaminoglycans, was upregulated, while type I collagen was downregulated in the dermis of DM and SLE skin. Interferon stimulated PSMB9 expression in cultured keratinocytes and reduced collagen expression in dermal fibroblasts, but did not affect versican expression. The PSMB9 knock-down in keratinocytes led to significant suppression of transforming growth factor (TGF)-ß2 and TGF-ß3, inducers of versican synthesis. TGF-ß3 expression was upregulated in both DM and SLE, while TGF-ß2 expression was increased only in the DM epidermis. ΔDiHS-diS1, a component of heparan sulfate, was significantly increased only in DM. TGF-ß2 expression significantly increased the ΔDiHS-diS1 expression in dermal fibroblasts in vitro. CONCLUSIONS: The interferon signature in DM and SLE skin reduces collagen in dermal fibroblasts, whereas overexpression of PSMB9 induced by interferon stimulates versican inducers in epidermal keratinocytes. In addition, the TGF-ß2-ΔDiHS-diS1 pathway may be responsible for the specific molecular change in DM skin.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Fármacos Dermatológicos/farmacología , Dermatomiositis/etiología , Interferones/farmacología , Lupus Eritematoso Sistémico/etiología , Colágeno Tipo I/metabolismo , Dermatomiositis/metabolismo , Femenino , Expresión Génica , Humanos , Queratinocitos/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Masculino , Persona de Mediana Edad , Proteómica , Piel/metabolismo , Factores de Crecimiento Transformadores/metabolismo , Regulación hacia Arriba/fisiología , Versicanos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA