Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 68(4): 645-658.e5, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29149593

RESUMEN

Hajdu-Cheney syndrome (HCS), a rare autosomal disorder caused by heterozygous mutations in NOTCH2, is clinically characterized by acro-osteolysis, severe osteoporosis, short stature, neurological symptoms, cardiovascular defects, and polycystic kidneys. Recent studies identified that aberrant NOTCH2 signaling and consequent osteoclast hyperactivity are closely associated with the bone-related disorder pathogenesis, but the exact molecular mechanisms remain unclear. Here, we demonstrate that sustained osteoclast activity is largely due to accumulation of NOTCH2 carrying a truncated C terminus that escapes FBW7-mediated ubiquitination and degradation. Mice with osteoclast-specific Fbw7 ablation revealed osteoporotic phenotypes reminiscent of HCS, due to elevated Notch2 signaling. Importantly, administration of Notch inhibitors in Fbw7 conditional knockout mice alleviated progressive bone resorption. These findings highlight the molecular basis of HCS pathogenesis and provide clinical insights into potential targeted therapeutic strategies for skeletal disorders associated with the aberrant FBW7/NOTCH2 pathway as observed in patients with HCS.


Asunto(s)
Proteína 7 que Contiene Repeticiones F-Box-WD , Síndrome de Hajdu-Cheney , Mutación , Osteoporosis , Proteolisis , Receptor Notch2 , Animales , Línea Celular , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Síndrome de Hajdu-Cheney/genética , Síndrome de Hajdu-Cheney/metabolismo , Ratones Noqueados , Osteoporosis/genética , Osteoporosis/metabolismo , Receptor Notch2/genética , Receptor Notch2/metabolismo , Ubiquitinación/genética
2.
FASEB J ; 34(11): 14930-14945, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32931083

RESUMEN

Proteasome inhibitors exert an anabolic effect on bone formation with elevated levels of osteoblast markers. These findings suggest the important role of the proteasomal degradation of osteogenic regulators, while the underlying molecular mechanisms are not fully understood. Here, we report that the proteasome inhibitors bortezomib and ixazomib markedly increased protein levels of the osteoblastic key transcription factor osterix/Sp7 (Osx). Furthermore, we revealed that Osx was targeted by p38 and Fbw7 for proteasomal degradation. Mechanistically, p38-mediated Osx phosphorylation at S73/77 facilitated Fbw7 interaction to trigger subsequent Osx ubiquitination. Consistent with these findings, p38 knockdown or pharmacological p38 inhibition resulted in Osx protein stabilization. Treatment with p38 inhibitors following osteogenic stimulation efficiently induced osteoblast differentiation through Osx stabilization. Conversely, pretreatment of p38 inhibitor followed by osteogenic challenge impaired osteoblastogenesis via suppressing Osx expression, suggesting that p38 exerts dual but opposite effects in the regulation of Osx level to fine-tune its activity during osteoblast differentiation. Furthermore, Fbw7-depleted human mesenchymal stem cells and primary mouse calvarial cells resulted in increased osteogenic capacity. Together, our findings unveil the molecular mechanisms underlying the Osx protein stability control and suggest that targeting the Osx degradation pathway could help enhance efficient osteogenesis and bone matrix regeneration.


Asunto(s)
Diferenciación Celular , Osteoblastos/metabolismo , Proteolisis , Factor de Transcripción Sp7/metabolismo , Animales , Compuestos de Boro/farmacología , Bortezomib/farmacología , Células Cultivadas , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Glicina/análogos & derivados , Glicina/farmacología , Células HCT116 , Células HEK293 , Humanos , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Factor de Transcripción Sp7/genética , Ubiquitinación , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Inflamm Res ; 70(5): 539-541, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33811487

RESUMEN

OBJECTIVE: Histamine derived from mast cells and basophils plays important roles in inducing allergic symptoms. Although T cells also produce histamine, the involvement of the histamine produced from T cells has remained enigmatic. We sought to reveal the roles of T helper 2 (Th2) cell-derived histamine in nasal allergic disorders. METHODS: The histamine production from Th2 cells was measured by EIA. The mRNA expression of histidine decarboxylase (HDC) was measured by real-time PCR. To investigate the roles of Th2 cell-derived histamine in vivo, we analyzed an antigen-specific Th2 cell transfer mouse model. RESULTS: Th2 cells produced histamine by T cell receptor stimulation, and these properties were specific for Th2 cells, but not Th1 cells and naïve CD4 T cells. The histamine produced from Th2 cells was involved in the infiltrations of Th2 cells in response to antigen exposure. CONCLUSION: These results suggest that Th2 cell-derived histamine play important roles in nasal allergic disorders.


Asunto(s)
Histamina/inmunología , Mucosa Nasal/inmunología , Rinitis Alérgica/inmunología , Células Th2/inmunología , Alérgenos/inmunología , Animales , Movimiento Celular , Histidina Descarboxilasa/genética , Ratones Endogámicos BALB C , Ratones Noqueados , Ovalbúmina/inmunología , Células Th2/fisiología , Células Th2/trasplante
4.
J Biol Chem ; 294(38): 14135-14148, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31366726

RESUMEN

The tumor suppressor CYLD is a deubiquitinating enzyme that suppresses polyubiquitin-dependent signaling pathways, including the proinflammatory and cell growth-promoting NF-κB pathway. Missense mutations in the CYLD gene are present in individuals with syndromes such as multiple familial trichoepithelioma (MFT), but the pathogenic roles of these mutations remain unclear. Recent studies have shown that CYLD interacts with a RING finger domain protein, mind bomb homologue 2 (MIB2), in the regulation of NOTCH signaling. However, whether MIB2 is an E3 ubiquitin ligase that acts on CYLD is unknown. Here, using the cell-free-based AlphaScreen and pulldown assays to detect protein-protein interactions, along with immunofluorescence assays and murine Mib2 knockout cells and animals, we demonstrate that MIB2 promotes proteasomal degradation of CYLD and enhances NF-κB signaling. Of note, arthritic inflammation was suppressed in Mib2-deficient mice. We further observed that the ankyrin repeat in MIB2 interacts with the third CAP domain in CYLD and that MIB2 catalyzes Lys-48-linked polyubiquitination of CYLD at Lys-338 and Lys-530. MIB2-dependent CYLD degradation activated NF-κB signaling via tumor necrosis factor alpha (TNFα) stimulation and the linear ubiquitination assembly complex (LUBAC). Mib2-knockout mice had reduced serum interleukin-6 (IL-6) and exhibited suppressed inflammatory responses in the K/BxN serum-transfer arthritis model. Interestingly, MIB2 significantly enhanced the degradation of a CYLDP904L variant identified in an individual with MFT, although the molecular pathogenesis of the disease was not clarified here. Together, these results suggest that MIB2 enhances NF-κB signaling in inflammation by promoting the ubiquitin-dependent degradation of CYLD.


Asunto(s)
Enzima Desubiquitinante CYLD/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Cisteína Endopeptidasas/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Femenino , Células HEK293 , Células HeLa , Humanos , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal/fisiología , Factor de Transcripción ReIA , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/metabolismo , Ubiquitinación
5.
Biochem Biophys Res Commun ; 524(2): 477-483, 2020 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-32008742

RESUMEN

Lipin-2 is a phosphatidate phosphatase with key roles in regulating lipid storage and energy homeostasis. LPIN2-genetic deficiency is associated with an autoinflammatory disorder, underscoring its critical role in innate immune signaling; however, the regulatory mechanisms underlying protein stability remain unknown. Here, we demonstrate that Lipin-2 interacts with ß-TRCP, a substrate receptor subunit of the SCFß-TRCP E3 ligase, and undergoes ubiquitination and proteasomal degradation. ß-TRCP-knockout in RAW264.7 macrophages resulted in Lipin-2 accumulation, leading to the suppression of LPS-induced MAPK activation and subsequent proinflammatory gene expression. Consistent with this, treatment with MLN4924, a Cullin-neddylation inhibitor that suppresses SCF E3 activity, increased Lipin-2 protein and concomitantly decreased Il1b expression. These findings suggested that ß-TRCP-mediated Lipin-2 degradation affects macrophage-elicited proinflammatory responses and could lead to new therapeutic approaches to treat inflammatory diseases.


Asunto(s)
Inflamación/metabolismo , Macrófagos/metabolismo , Fosfatidato Fosfatasa/metabolismo , Proteolisis , Animales , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Inflamación/genética , Ratones , Fosfatidato Fosfatasa/genética , Células RAW 264.7 , Ubiquitinación , Proteínas con Repetición de beta-Transducina/genética , Proteínas con Repetición de beta-Transducina/metabolismo
6.
Mol Biol Evol ; 31(12): 3282-301, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25205508

RESUMEN

The caspases, a family of cysteine proteases, play multiple roles in apoptosis, inflammation, and cellular differentiation. Caspase-8 (Casp8), which was first identified in humans, functions as an initiator caspase in the apoptotic signaling mediated by cell-surface death receptors. To understand the evolution of function in the Casp8 protein family, casp8 orthologs were identified from a comprehensive range of vertebrates and invertebrates, including sponges and cnidarians, and characterized at both the gene and protein levels. Some introns have been conserved from cnidarians to mammals, but both losses and gains have also occurred; a new intron arose during teleost evolution, whereas in the ascidian Ciona intestinalis, the casp8 gene is intronless and is organized in an operon with a neighboring gene. Casp8 activities are near ubiquitous throughout the animal kingdom. Exogenous expression of a representative range of nonmammalian Casp8 proteins in cultured mammalian cells induced cell death, implying that these proteins possess proapoptotic activity. The cnidarian Casp8 proteins differ considerably from their bilaterian counterparts in terms of amino acid residues in the catalytic pocket, but display the same substrate specificity as human CASP8, highlighting the complexity of spatial structural interactions involved in enzymatic activity. Finally, it was confirmed that the interaction with an adaptor molecule, Fas-associated death domain protein, is also evolutionarily ancient. Thus, despite structural diversity and cooption to a variety of new functions, the ancient origins and near ubiquitous distribution of this activity across the animal kingdom emphasize the importance and utility of Casp8 as a central component of the metazoan molecular toolkit.


Asunto(s)
Apoptosis , Caspasa 8/genética , Secuencia de Aminoácidos , Animales , Anélidos/genética , Antozoos/genética , Secuencia de Bases , Caspasa 8/química , Ciona intestinalis/genética , Evolución Molecular , Proteínas de Peces/genética , Peces/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Mytilus/genética , Filogenia , Planarias/genética , Conformación Proteica , Especificidad por Sustrato
7.
Antioxidants (Basel) ; 12(2)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36829909

RESUMEN

Protein ubiquitination, which is catalyzed by ubiquitin-activating enzymes, ubiquitin-conjugating enzymes, and ubiquitin ligases, is a crucial post-translational modification to regulate numerous cellular functions in a spatio-temporal-specific manner. The human genome encodes ~100 deubiquitinating enzymes (DUBs), which antagonistically regulate the ubiquitin system. OTUD1, an ovarian tumor protease (OTU) family DUB, has an N-terminal-disordered alanine-, proline-, glycine-rich region (APGR), a catalytic OTU domain, and a ubiquitin-interacting motif (UIM). OTUD1 preferentially hydrolyzes lysine-63-linked ubiquitin chains in vitro; however, recent studies indicate that OTUD1 cleaves various ubiquitin linkages, and is involved in the regulation of multiple cellular functions. Thus, OTUD1 predominantly functions as a tumor suppressor by targeting p53, SMAD7, PTEN, AKT, IREB2, YAP, MCL1, and AIF. Furthermore, OTUD1 regulates antiviral signaling, innate and acquired immune responses, and cell death pathways. Similar to Nrf2, OTUD1 contains a KEAP1-binding ETGE motif in its APGR and regulates the reactive oxygen species (ROS)-mediated oxidative stress response and cell death. Importantly, in addition to its association with various cancers, including multiple myeloma, OTUD1 is involved in acute graft-versus-host disease and autoimmune diseases such as systemic lupus erythematosus, rheumatoid arthritis, and ulcerative colitis. Thus, OTUD1 is an important DUB as a therapeutic target for a variety of diseases.

8.
Front Mol Biosci ; 10: 1089213, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36726375

RESUMEN

In neurodegenerative diseases such as Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS), the progressive accumulation of ubiquitin-positive cytoplasmic inclusions leads to proteinopathy and neurodegeneration. Along with the seven types of Lys-linked ubiquitin chains, the linear ubiquitin chain assembly complex (LUBAC)-mediated Met1-linked linear ubiquitin chain, which activates the canonical NF-κB pathway, is also involved in cytoplasmic inclusions of tau in AD and TAR DNA-binding protein 43 in ALS. Post-translational modifications, including heterologous ubiquitination, affect proteasomal and autophagic degradation, inflammatory responses, and neurodegeneration. Single nucleotide polymorphisms (SNPs) in SHARPIN and RBCK1 (which encodes HOIL-1L), components of LUBAC, were recently identified as genetic risk factors of AD. A structural biological simulation suggested that most of the SHARPIN SNPs that cause an amino acid replacement affect the structure and function of SHARPIN. Thus, the aberrant LUBAC activity is related to AD. Protein ubiquitination and ubiquitin-binding proteins, such as ubiquilin 2 and NEMO, facilitate liquid-liquid phase separation (LLPS), and linear ubiquitination seems to promote efficient LLPS. Therefore, the development of therapeutic approaches that target ubiquitination, such as proteolysis-targeting chimeras (PROTACs) and inhibitors of ubiquitin ligases, including LUBAC, is expected to be an additional effective strategy to treat neurodegenerative diseases.

9.
Cell Death Dis ; 13(8): 694, 2022 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-35941131

RESUMEN

Deubiquitinating enzymes (DUBs) regulate numerous cellular functions by removing ubiquitin modifications. We examined the effects of 88 human DUBs on linear ubiquitin chain assembly complex (LUBAC)-induced NF-κB activation, and identified OTUD1 as a potent suppressor. OTUD1 regulates the canonical NF-κB pathway by hydrolyzing K63-linked ubiquitin chains from NF-κB signaling factors, including LUBAC. OTUD1 negatively regulates the canonical NF-κB activation, apoptosis, and necroptosis, whereas OTUD1 upregulates the interferon (IFN) antiviral pathway. Mass spectrometric analysis showed that OTUD1 binds KEAP1, and the N-terminal intrinsically disordered region of OTUD1, which contains an ETGE motif, is indispensable for the KEAP1-binding. Indeed, OTUD1 is involved in the KEAP1-mediated antioxidant response and reactive oxygen species (ROS)-induced cell death, oxeiptosis. In Otud1-/--mice, inflammation, oxidative damage, and cell death were enhanced in inflammatory bowel disease, acute hepatitis, and sepsis models. Thus, OTUD1 is a crucial regulator for the inflammatory, innate immune, and oxidative stress responses and ROS-associated cell death pathways.


Asunto(s)
Factor 2 Relacionado con NF-E2 , FN-kappa B , Animales , Muerte Celular , Enzimas Desubicuitinizantes/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ubiquitina/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitinación
10.
PLoS One ; 16(3): e0248158, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33662037

RESUMEN

Histamine, which is mainly produced by mast cells and basophils, participates in various allergic symptoms, and some studies have reported that macrophages also produce histamine. Moreover, recent studies have revealed that macrophages, especially alternatively activated macrophages (M2) induced by T helper 2 (Th2) cytokines, such as interleukin (IL)-4 and IL-13, participate in the pathogenesis of allergic diseases. The major source of Th2 cytokines is antigen-specific Th2 cells. To elucidate the relationship between histamine, macrophages, and Th2 cells in allergic inflammation, we established a macrophage-Th2 cell co-culture model in vitro and an antigen-specific Th2 cell transfer mouse model of rhinitis. In vitro analyses indicated that macrophages produce histamine by interacting with antigen-specific Th2 cells through the antigen. Furthermore, Th2 cells and macrophages cooperatively elicited rhinitis in the mouse model. We determined that histamine induces Th2- and macrophage-elicited sneezing responses through H1 receptor signaling, whereas it induces nasal eosinophil infiltrations through H4 receptor signaling. Collectively, these results indicate a novel histamine production mechanism by macrophages, in which Th2 cells and macrophages cooperatively induce nasal allergic inflammation through histamine signaling.


Asunto(s)
Histamina/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Rinitis Alérgica/inmunología , Células Th2/inmunología , Animales , Células Cultivadas , Humanos , Inflamación/patología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Rinitis Alérgica/patología , Transducción de Señal , Células Th2/patología
11.
Cell Rep ; 37(6): 109988, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758305

RESUMEN

The anti-apoptotic myeloid cell leukemia 1 (MCL1) protein belongs to the pro-survival BCL2 family and is frequently amplified or elevated in human cancers. MCL1 is highly unstable, with its stability being regulated by phosphorylation and ubiquitination. Here, we identify acetylation as another critical post-translational modification regulating MCL1 protein stability. We demonstrate that the lysine acetyltransferase p300 targets MCL1 at K40 for acetylation, which is counteracted by the deacetylase sirtuin 3 (SIRT3). Mechanistically, acetylation enhances MCL1 interaction with USP9X, resulting in deubiquitination and subsequent MCL1 stabilization. Therefore, ectopic expression of acetylation-mimetic MCL1 promotes apoptosis evasion of cancer cells, enhances colony formation potential, and facilitates xenografted tumor progression. We further demonstrate that elevated MCL1 acetylation sensitizes multiple cancer cells to pharmacological inhibition of USP9X. These findings reveal that acetylation of MCL1 is a critical post-translational modification enhancing its oncogenic function and provide a rationale for developing innovative therapeutic strategies for MCL1-dependent tumors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Neoplasias/patología , Estabilidad Proteica , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Células Tumorales Cultivadas , Ubiquitina Tiolesterasa/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Factores de Transcripción p300-CBP/genética
12.
Nat Commun ; 12(1): 5386, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34508104

RESUMEN

Although inhibitors targeting CDK4/6 kinases (CDK4/6i) have shown promising clinical prospect in treating ER+/HER2- breast cancers, acquired drug resistance is frequently observed and mechanistic knowledge is needed to harness their full clinical potential. Here, we report that inhibition of CDK4/6 promotes ßTrCP1-mediated ubiquitination and proteasomal degradation of RB1, and facilitates SP1-mediated CDK6 transcriptional activation. Intriguingly, suppression of CK1ε not only efficiently prevents RB1 from degradation, but also prevents CDK4/6i-induced CDK6 upregulation by modulating SP1 protein stability, thereby enhancing CDK4/6i efficacy and overcoming resistance to CDK4/6i in vitro. Using xenograft and PDX models, we further demonstrate that combined inhibition of CK1ε and CDK4/6 results in marked suppression of tumor growth in vivo. Altogether, these results uncover the molecular mechanisms by which CDK4/6i treatment alters RB1 and CDK6 protein abundance, thereby driving the acquisition of CDK4/6i resistance. Importantly, we identify CK1ε as an effective target for potentiating the therapeutic efficacy of CDK4/6 inhibitors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Caseína Cinasa 1 épsilon/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Proteínas Quinasas/uso terapéutico , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteínas de Unión a Retinoblastoma/metabolismo , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Influenza Other Respir Viruses ; 15(4): 488-494, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33715290

RESUMEN

BACKGROUND: Coronavirus disease (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was first detected in Japan in January 2020 and has spread throughout the country. Previous studies have reported that viral interference among influenza virus, rhinovirus, and other respiratory viruses can affect viral infections at the host and population level. METHODS: To investigate the impact of COVID-19 on influenza and other respiratory virus infections, we analyzed clinical specimens collected from 2244 patients in Japan with respiratory diseases between January 2018 and September 2020. RESULTS: The frequency of influenza and other respiratory viruses (coxsackievirus A and B; echovirus; enterovirus; human coronavirus 229E, HKU1, NL63, and OC43; human metapneumovirus; human parainfluenza virus 1, 2, 3, and 4; human parechovirus; human respiratory syncytial virus; human adenovirus; human bocavirus; human parvovirus B19; herpes simplex virus type 1; and varicella-zoster virus) was appreciably reduced among all patients during the COVID-19 pandemic except for that of rhinovirus in children younger than 10 years, which was appreciably increased. COVID-19 has not spread among this age group, suggesting an increased risk of rhinovirus infection in children. CONCLUSIONS: Rhinovirus infections should be continuously monitored to understand their increased risk during the COVID-19 pandemic and viral interference with SARS-CoV-2.


Asunto(s)
COVID-19/epidemiología , Infecciones por Picornaviridae/epidemiología , Rhinovirus/aislamiento & purificación , Adulto , Niño , Preescolar , Coinfección/diagnóstico , Coinfección/epidemiología , Coinfección/virología , Femenino , Humanos , Lactante , Recién Nacido , Japón/epidemiología , Masculino , Infecciones por Picornaviridae/diagnóstico , Infecciones por Picornaviridae/virología , Infecciones del Sistema Respiratorio/diagnóstico , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Riesgo , SARS-CoV-2 , Virosis/diagnóstico , Virosis/epidemiología , Virosis/virología , Virus/aislamiento & purificación
14.
BMC Biotechnol ; 10: 42, 2010 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-20525307

RESUMEN

BACKGROUND: Technology used for the purification of recombinant proteins is a key issue for the biochemical and structural analyses of proteins. In general, affinity tags, such as glutathione-S-transferase or six-histidines, are used to purify recombinant proteins. Since such affinity tags often interfere negatively with the structural and functional analyses of proteins, they are usually removed by treatment with proteases. Previously, Dr. H. Mao reported self-cleavage purification of a target protein by fusing the sortase protein to its N-terminal end, and subsequently obtained tag-free recombinant protein following expression in Escherichia coli. This method, however, is yet to be applied to the cell-free based protein production. RESULTS: The histidine tag-based self-cleavage method for purifying proteins produced by the wheat cell-free protein synthesis system showed high background, low recovery, and unexpected cleavage between the N-terminally fused sortase and target protein during the protein synthesis. Addition of calcium chelator BAPTA to the cell-free reaction inhibited the cleavage. In order to adapt the sortase-based purification method to the cell-free system, we next used biotin as the affinity tag. The biotinylated sortase self-cleavage purification (BISOP) method provided tag-free, highly purified proteins due to improved recovery of proteins from the resin. The N-terminal sequence analysis of the GFP produced by the BISOP method revealed that the cleavage indeed occurred at the right cleavage site. Using this method, we also successfully purified the E2 heterocomplex of USE2N and USE2v1. The c-terminal src kinase (CSK) obtained by the BISOP method showed high activity in phosphorylating the Src protein. Furthermore, we demonstrated that this method is suitable for automatically synthesizing and purifying proteins using robots. CONCLUSION: We demonstrated that the newly developed BISOP method is very useful for obtaining high quality, tag-free recombinant proteins, produced using the cell-free system, for biochemical and structural analyses.


Asunto(s)
Biotinilación/métodos , Proteínas/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Sistema Libre de Células , Clonación Molecular , Fosforilación , Plásmidos
15.
Cell Signal ; 46: 15-22, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29474981

RESUMEN

FBW7 is one of the most well characterized F-box proteins that serve as substrate recognition subunits of SCF (Skp1-Cullin 1-F-box proteins) E3 ubiquitin ligase complexes. SCFFBW7 plays key roles in regulating cell cycle progression, differentiation, and stem cell maintenance largely through targeting a broad range of oncogenic substrates for proteasome-dependent degradation. The identification of an increasing number of FBW7 substrates for ubiquitination, and intensive in vitro and in vivo studies have revealed a network of signaling components controlled by FBW7 that contributes to metabolic regulation as well as its tumor suppressor role. Here we mainly focus on recent findings that highlight a critical role for FBW7 in cancer and metabolism.


Asunto(s)
Proteína 7 que Contiene Repeticiones F-Box-WD/fisiología , Neoplasias/metabolismo , Ubiquitinación , Animales , Ciclo Celular , Diferenciación Celular , Línea Celular Tumoral , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Humanos , Ratones , Fosforilación , Transducción de Señal , Proteínas Supresoras de Tumor/fisiología
16.
Sci Signal ; 10(466)2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28196907

RESUMEN

Abnormal activation of the oncogenic E3 ubiquitin ligase murine double minute 2 (MDM2) is frequently observed in human cancers. By ubiquitinating the tumor suppressor p53 protein, which leads to its proteasome-mediated destruction, MDM2 limits the tumor-suppressing activity of p53. On the other hand, by ubiquitinating itself, MDM2 targets itself for destruction and promotes the p53 tumor suppressor pathway, a process that can be antagonized by the deubiquitinase herpesvirus-associated ubiquitin-specific protease (HAUSP). We investigated the regulation of MDM2 substrate specificity and found that acetyltransferase p300-mediated acetylation and stabilization of MDM2 are molecular switches that block self-ubiquitination, thereby shifting its E3 ligase activity toward p53. In vitro and in cancer cell lines, p300-mediated acetylation of MDM2 on Lys182 and Lys185 enabled HAUSP to bind, presumably deubiquitinate, and stabilize MDM2. This acetylation within the nuclear localization signal domain decreased its interaction with the acidic domain, subsequently increased the interaction between the acidic domain and RING domain in MDM2, enabled the binding of HAUSP to the acidic domain in MDM2, and shifted MDM2 activity from autoubiquitination to p53 ubiquitination. However, upon genotoxic stress through exposure to etoposide, the deacetylase sirtuin 1 (SIRT1) deacetylated MDM2 at Lys182 and Lys185, thereby promoting self-ubiquitination and less ubiquitination and subsequent degradation of p53, thus increasing p53-dependent apoptosis. Therefore, this study indicates that dynamic acetylation is a molecular switch in the regulation of MDM2 substrate specificity, revealing further insight into the posttranslational regulation of the MDM2/p53 cell survival axis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Oncogenes , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Células A549 , Acetilación , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células HCT116 , Células HEK293 , Humanos , Immunoblotting , Lisina/genética , Lisina/metabolismo , Células MCF-7 , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/genética , Interferencia de ARN , Sirtuina 1/genética , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Peptidasa Específica de Ubiquitina 7/metabolismo , Ubiquitinación , Factores de Transcripción p300-CBP/genética
17.
Oncotarget ; 8(6): 9947-9960, 2017 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-28039480

RESUMEN

Folliculin-interacting protein 1 and 2 (FNIP1 and FNIP2) play critical roles in preventing renal malignancy through their association with the tumor suppressor FLCN. Mutations in FLCN are associated with Birt-Hogg-Dubé (BHD) syndrome, a rare disorder with increased risk of renal cancer. Recent studies indicated that FNIP1/FNIP2 double knockout mice display enlarged polycystic kidneys and renal carcinoma, which phenocopies FLCN knockout mice, suggesting that these two proteins function together to suppress renal cancer. However, the molecular mechanism functionally linking FNIP1/FNIP2 and FLCN remains largely elusive. Here, we demonstrated that FNIP2 protein is unstable and subjected to proteasome-dependent degradation via ß-TRCP and Casein Kinase 1 (CK1)-directed ubiquitination in a nutrition-dependent manner. Degradation of FNIP2 leads to lysosomal dissociation of FLCN and subsequent lysosomal association of mTOR, which in turn promotes the proliferation of renal cancer cells. These results indicate that SCFß-TRCP negatively regulates the FLCN complex by promoting FNIP degradation and provide molecular insight into the pathogenesis of BHD-associated renal cancer.


Asunto(s)
Síndrome de Birt-Hogg-Dubé/enzimología , Carcinoma de Células Renales/enzimología , Proteínas Portadoras/metabolismo , Proliferación Celular , Neoplasias Renales/enzimología , Estado Nutricional , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Síndrome de Birt-Hogg-Dubé/genética , Síndrome de Birt-Hogg-Dubé/patología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Proteínas Portadoras/genética , Quinasa de la Caseína I/metabolismo , Metabolismo Energético , Células HEK293 , Células HeLa , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Lisosomas/metabolismo , Ratones Desnudos , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteolisis , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Proteínas Ligasas SKP Cullina F-box/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral , Proteínas Supresoras de Tumor/genética , Ubiquitinación
18.
Nat Cell Biol ; 19(3): 177-188, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28192421

RESUMEN

Progression of mammalian cells through the G1 and S phases of the cell cycle is driven by the D-type and E-type cyclins. According to the current models, at least one of these cyclin families must be present to allow cell proliferation. Here, we show that several cell types can proliferate in the absence of all G1 cyclins. However, following ablation of G1 cyclins, embryonic stem (ES) cells attenuated their pluripotent characteristics, with the majority of cells acquiring the trophectodermal cell fate. We established that G1 cyclins, together with their associated cyclin-dependent kinases (CDKs), phosphorylate and stabilize the core pluripotency factors Nanog, Sox2 and Oct4. Treatment of murine ES cells, patient-derived glioblastoma tumour-initiating cells, or triple-negative breast cancer cells with a CDK inhibitor strongly decreased Sox2 and Oct4 levels. Our findings suggest that CDK inhibition might represent an attractive therapeutic strategy by targeting glioblastoma tumour-initiating cells, which depend on Sox2 to maintain their tumorigenic potential.


Asunto(s)
Diferenciación Celular , Ciclina G1/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Animales , Biomarcadores/metabolismo , Ciclo Celular , Proliferación Celular , Separación Celular , ADN/análisis , Embrión de Mamíferos/citología , Epigénesis Genética , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Histonas/metabolismo , Hormonas/farmacología , Imagenología Tridimensional , Lisina/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Metilación , Ratones , Ratones Endogámicos C57BL , ARN/análisis , Receptores Acoplados a Proteínas G/metabolismo , Esteroides/farmacología , Tetraspaninas/metabolismo
19.
Sci Signal ; 10(460)2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28049764

RESUMEN

The SCFß-TRCP E3 ubiquitin ligase complex plays pivotal roles in normal cellular physiology and in pathophysiological conditions. Identification of ß-transducin repeat-containing protein (ß-TRCP) substrates is therefore critical to understand SCFß-TRCP biology and function. We used a ß-TRCP-phosphodegron motif-specific antibody in a ß-TRCP substrate screen coupled with tandem mass spectrometry and identified multiple ß-TRCP substrates. One of these substrates was Lipin1, an enzyme and suppressor of the family of sterol regulatory element-binding protein (SREBP) transcription factors, which activate genes encoding lipogenic factors. We showed that SCFß-TRCP specifically interacted with and promoted the polyubiquitination of Lipin1 in a manner that required phosphorylation of Lipin1 by mechanistic target of rapamycin 1 (mTORC1) and casein kinase I (CKI). ß-TRCP depletion in HepG2 hepatocellular carcinoma cells resulted in increased Lipin1 protein abundance, suppression of SREBP-dependent gene expression, and attenuation of triglyceride synthesis. Moreover, ß-TRCP1 knockout mice showed increased Lipin1 protein abundance and were protected from hepatic steatosis induced by a high-fat diet. Together, these data reveal a critical physiological function of ß-TRCP in regulating hepatic lipid metabolic homeostasis in part through modulating Lipin1 stability.


Asunto(s)
Lipogénesis , Hígado/metabolismo , Proteínas Nucleares/metabolismo , Fosfatidato Fosfatasa/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Immunoblotting , Ratones , Ratones Noqueados , Células 3T3 NIH , Proteínas Nucleares/genética , Fosfatidato Fosfatasa/genética , Fosforilación , Unión Proteica , Proteolisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ligasas SKP Cullina F-box/genética , Especificidad por Sustrato , Ubiquitinación
20.
Nat Med ; 23(9): 1063-1071, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28805820

RESUMEN

The bromodomain and extraterminal (BET) family of proteins comprises four members-BRD2, BRD3, BRD4 and the testis-specific isoform BRDT-that largely function as transcriptional coactivators and play critical roles in various cellular processes, including the cell cycle, apoptosis, migration and invasion. BET proteins enhance the oncogenic functions of major cancer drivers by elevating the expression of these drivers, such as c-Myc in leukemia, or by promoting the transcriptional activities of oncogenic factors, such as AR and ERG in prostate cancer. Pathologically, BET proteins are frequently overexpressed and are clinically linked to various types of human cancer; they are therefore being pursued as attractive therapeutic targets for selective inhibition in patients with cancer. To this end, a number of bromodomain inhibitors, including JQ1 and I-BET, have been developed and have shown promising outcomes in early clinical trials. Although resistance to BET inhibitors has been documented in preclinical models, the molecular mechanisms underlying acquired resistance are largely unknown. Here we report that cullin-3SPOP earmarks BET proteins, including BRD2, BRD3 and BRD4, for ubiquitination-mediated degradation. Pathologically, prostate cancer-associated SPOP mutants fail to interact with and promote the degradation of BET proteins, leading to their elevated abundance in SPOP-mutant prostate cancer. As a result, prostate cancer cell lines and organoids derived from individuals harboring SPOP mutations are more resistant to BET-inhibitor-induced cell growth arrest and apoptosis. Therefore, our results elucidate the tumor-suppressor role of SPOP in prostate cancer in which it acts as a negative regulator of BET protein stability and also provide a molecular mechanism for resistance to BET inhibitors in individuals with prostate cancer bearing SPOP mutations.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/genética , Proteínas Represoras/genética , Factores de Transcripción/metabolismo , Apoptosis , Azepinas , Benzodiazepinas , Proteínas de Ciclo Celular , Proliferación Celular , Proteínas Cullin , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Masculino , Terapia Molecular Dirigida , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas de Unión al ARN , Talidomida/análogos & derivados , Triazoles , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA