Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
Mol Cell Endocrinol ; 592: 112283, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38815795

RESUMEN

Adipose tissue macrophages (ATMs) are key players in the development of obesity and associated metabolic inflammation, which contributes to systemic metabolic dysfunction, and understanding the interaction between macrophages and adipocytes is crucial for developing novel macrophage-based strategies against obesity. Here, we found that Legumain (Lgmn), a well-known lysosomal cysteine protease, is expressed mainly in the ATMs of obese mice. To further define the potential role of Lgmn-expressing macrophages in the generation of an aberrant metabolic state, LgmnF/F; LysMCre mice, which do not express Lgmn in macrophages, were maintained on a high-fat diet (HFD), and metabolic parameters were assessed. Macrophage-specific Lgmn deficiency protects mice against HFD-induced obesity, diminishes the quantity of proinflammatory macrophages in obese adipose tissues, and alleviates hepatic steatosis and insulin resistance. By analysing the transcriptome and proteome of murine visceral white adipose tissue (vWAT) after HFD feeding, we determined that macrophage Lgmn deficiency causes changes in lipid metabolism and the inflammatory response. Furthermore, the reciprocity of macrophage-derived Lgmn with integrin α5ß1 in adipocytes was tested via colocalization analyses. It is further demonstrated in macrophage and adipocyte coculture system that macrophage derived Lgmn bound to integrin α5ß1 in adipocytes, therefore attenuating PKA activation, downregulating lipolysis-related proteins and eventually exacerbating obesity development. Overall, our study identified Lgmn as a previously unrecognized regulator involved in the interaction between ATMs and adipocytes contributing to diet-induced obesity and suggested that Lgmn is a potential target for treating metabolic disorders.


Asunto(s)
Tejido Adiposo , Cisteína Endopeptidasas , Dieta Alta en Grasa , Inflamación , Metabolismo de los Lípidos , Macrófagos , Ratones Endogámicos C57BL , Obesidad , Animales , Obesidad/metabolismo , Obesidad/genética , Macrófagos/metabolismo , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/deficiencia , Dieta Alta en Grasa/efectos adversos , Inflamación/patología , Inflamación/metabolismo , Inflamación/genética , Ratones , Tejido Adiposo/metabolismo , Adipocitos/metabolismo , Masculino , Resistencia a la Insulina
2.
Behav Brain Res ; 413: 113464, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34265317

RESUMEN

Emotional and cognitive impairment has been recognized as a central feature of depression, which is closely related to hyperfunction of the hypothalamic-pituitary-adrenal (HPA) axis caused by down-regulation of glucocorticoid receptor (GR) expression in patients. A decrease in GR expression can cause pathological changes and lead to the impairment of synaptic plasticity. Legumain, a lysosomal cysteine protease, plays an important role in neurological diseases. It is reported that legumain activates the MAPK signaling pathway, which modifies the GR. Therefore, we hypothesize that regulation of the GR by legumain plays a crucial role in the pathological process of depression. The relationships between legumain, GR, synaptic plasticity and emotional and cognitive deficits were explored in this study. The results demonstrated that repeated corticosterone (CORT) injections (3 weeks) induced emotional and cognitive deficits in mice, based on behavioral experiments and the detection of synaptic plasticity. Furthermore, CORT injections decreased the expression of hippocampal synapse-related proteins, cell density and dendritic spine density in the hippocampus, accompanied by increased protein expression in the MAPK signaling pathway and decreased expression of the GR. In conclusion, our results demonstrated that legumain knockout up-regulated expression of the GR by reducing protein expression in the MAPK signaling pathway, thereby improving hippocampal synaptic plasticity as well as the emotional and cognitive impairment of model mice. This suggests that legumain may be an effective therapeutic target for emotional and cognitive deficits.


Asunto(s)
Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/enzimología , Corticosterona/farmacología , Cisteína Endopeptidasas/metabolismo , Depresión/inducido químicamente , Depresión/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Plasticidad Neuronal/fisiología , Animales , Conducta Animal/fisiología , Corticosterona/administración & dosificación , Cisteína Endopeptidasas/deficiencia , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Noqueados , Distribución Aleatoria
3.
Cancer Immunol Res ; 9(6): 682-692, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33707310

RESUMEN

Apart from the constitutive proteasome, the immunoproteasome that comprises the three proteolytic subunits LMP2, MECL-1, and LMP7 is expressed in most immune cells. In this study, we describe opposing roles for immunoproteasomes in regulating the tumor microenvironment (TME). During chronic inflammation, immunoproteasomes modulated the expression of protumorigenic cytokines and chemokines and enhanced infiltration of innate immune cells, thus triggering the onset of colitis-associated carcinogenesis (CAC) in wild-type mice. Consequently, immunoproteasome-deficient animals (LMP2/MECL-1/LMP7-null mice) were almost completely resistant to CAC development. In patients with ulcerative colitis with high risk for CAC, immunoproteasome-induced protumorigenic mediators were upregulated. In melanoma tumors, the role of immunoproteasomes is relatively unknown. We found that high expression of immunoproteasomes in human melanoma was associated with better prognosis. Similarly, our data revealed that the immunoproteasome has antitumorigenic activity in a mouse model of melanoma. The antitumor immunity against melanoma was compromised in immunoproteasome-deficient mice because of the impaired activity of CD8+ CTLs, CD4+ Th1 cells, and antigen-presenting cells. These findings show that immunoproteasomes may exert opposing roles with either pro- or antitumoral properties in a context-dependent manner.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Melanoma Experimental/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/inmunología , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Colitis/patología , Cisteína Endopeptidasas/deficiencia , Cisteína Endopeptidasas/genética , Citocinas/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Melanoma Experimental/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/genética , Linfocitos T Citotóxicos/metabolismo
4.
Clin Sci (Lond) ; 135(6): 811-828, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33687053

RESUMEN

Post-translational modification (PTM) by small ubiquitin-like modifier (SUMO) is a key regulator of cell proliferation and can be readily reversed by a family of SUMO-specific proteases (SENPs), making SUMOylation an ideal regulatory mechanism for developing novel therapeutic strategies for promoting a cardiac regenerative response. However, the role of SUMOylation in cardiac regeneration remains unknown. In the present study, we assessed whether targeting protein kinase B (Akt) SUMOylation can promote cardiac regeneration. Quantitative PCR and Western blotting results showed that small ubiquitin-like modifier-specific protease 2 (SENP2) is up-regulated during postnatal heart development. SENP2 deficiency promoted P7 and adult cardiomyocyte (CM) dedifferentiation and proliferation both in vitro and in vivo. Mice with SENP2 deficiency exhibited improved cardiac function after MI due to CM proliferation and angiogenesis. Mechanistically, the loss of SENP2 up-regulated Akt SUMOylation levels and increased Akt kinase activity, leading to a decrease in GSK3ß levels and subsequently promoting CM proliferation and angiogenesis. In summary, inhibition of SENP2-mediated Akt deSUMOylation promotes CM differentiation and proliferation by activating the Akt pathway. Our results provide new insights into the role of SUMOylation in cardiac regeneration.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/metabolismo , Sumoilación , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Cisteína Endopeptidasas/deficiencia , Cisteína Endopeptidasas/genética , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Ratones Endogámicos C57BL , Infarto del Miocardio , Miocitos Cardíacos/citología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regeneración/fisiología
5.
Mol Neurobiol ; 58(4): 1607-1620, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33219900

RESUMEN

Neuroinflammation is the important pathological feature of Alzheimer's disease (AD). Legumain, a lysosomal cysteine protease, plays an important role in neuroinflammation during ischemic stroke and depressive disorder. Legumain is involved in AD process through cleaving APP; however, it is unclear if legumain can possibly modulate neuroinflammation without cleaving APP in AD. Thus, we established a mouse model of AD by single intracerebroventricular injections of Aß1-42 in legumain knockout (KO) mice. The behavioral tests showed that legumain-KO effectively ameliorated cognitive impairment induced by Aß1-42. Moreover, legumain deprivation significantly improves the synaptic plasticity damages in Aß1-42-treated mice. Moreover, legumain-KO considerably inhibited the activation of microglia and reduced the expression of inflammatory cytokines in the hippocampus of Aß1-42-treated mice. Interestingly, we found that legumain-KO inhibited TLR4/MyD88/NF-κB pathway, which was activated by Aß1-42 in the hippocampus. In conclusion, our results suggested that legumain-KO reduced the level of neuroinflammation that was associated with inhibiting TLR4/MyD88/NF-κB pathways, thereby improving the hippocampal synaptic plasticity and reducing the cognitive impairments in Aß1-42-treated mice. Legumain knockout blocked microglia activation by inhibiting TLR4/MyD88/NF-κB signaling pathways, and further reduced inflammatory cytokine expression. As a result, legumain knockout alleviated synaptic damage and cognitive impairment induced by Aß1--42.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/toxicidad , Encéfalo/patología , Disfunción Cognitiva/complicaciones , Cisteína Endopeptidasas/deficiencia , Inflamación/patología , Plasticidad Neuronal , Fragmentos de Péptidos/toxicidad , Animales , Encéfalo/fisiopatología , Disfunción Cognitiva/fisiopatología , Cisteína Endopeptidasas/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Inflamación/complicaciones , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/fisiopatología , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Aprendizaje Espacial , Receptor Toll-Like 4/metabolismo
6.
Cell Biochem Biophys ; 78(4): 483-494, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32918681

RESUMEN

Tazarotene-induced gene 1 (TIG1) is a retinoid acid receptor-responsive gene involved in cell differentiation and tumorigenesis. Aberrant methylation of CpG islands in the TIG1 promoter is found in multiple cancers. Currently, the exact mechanism underlying the anticancer effect of TIG1 is unknown. Here, we show that TIG1 interacts with cathepsin V (CTSV), which reduces CTSV stability and subsequently affects the production of activated urokinase-type plasminogen activator (uPA), an epithelial-mesenchymal transition-associated protein. Ectopic expression of CTSV increased the expression of activated uPA and the number of migrated and invaded cells, whereas ectopic TIG1 expression reversed the effects of CTSV on the uPA signaling pathway. Similar patterns in the production of activated uPA and number of migrated and invaded cells were also observed in TIG1-expressing and CTSV-knockdown cells. The results suggest that CTSV may participate in TIG1-regulated uPA activity and the associated downstream signaling pathway.


Asunto(s)
Catepsinas/metabolismo , Neoplasias Colorrectales/patología , Cisteína Endopeptidasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Catepsinas/deficiencia , Catepsinas/genética , Movimiento Celular , Cisteína Endopeptidasas/deficiencia , Cisteína Endopeptidasas/genética , Transición Epitelial-Mesenquimal/genética , Silenciador del Gen , Células HCT116 , Humanos , Invasividad Neoplásica
7.
CNS Neurosci Ther ; 26(8): 815-828, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32495523

RESUMEN

AIMS: SUMOylation is a posttranslational modification related to multiple human diseases. SUMOylation can be reversed by classes of proteases known as the sentrin/SUMO-specific proteases (SENPs). In the present study, we investigate the potential role of SENP1 in pericytes in the brain ischemia. METHODS: Pericyte-specific deletion of senp1 mice (Cspg4-Cre; senp1f/f ) were used for brain function and neuronal damage evaluation following brain ischemia. The cerebral blood vessels of diameter, velocity, and flux were performed in living mice by two-photon laser scanning microscopy (TPLSM). Biochemical analysis and immunohistochemistry methods were used to address the role and mechanism of pericyte-specific SENP1 in the pathological process of brain ischemia. A coculture model of HBVPs and HBMECs mimicked the BBB in vitro and was used to evaluate BBB integrity after glucose deprivation. RESULTS: Our results showed that senp1-specific deletion in pericytes did not affect the motor function and cognitive function of mice. However, the pericyte-specific deletion of senp1 aggravated the infarct size and motor deficit following focal brain ischemia. Consistently, the TPLSM data demonstrated that SENP1 deletion in pericytes accelerated thrombosis formation in brain microvessels. We also found that pericyte-specific deletion of senp1 exaggerated the neuronal damage significantly following brain ischemia in mice. Moreover, SENP1 knockdown in pericytes could activate the apoptosis signaling and disrupt the barrier integrity in vitro coculture model. CONCLUSIONS: Our findings revealed that targeting SENP1 in pericytes may represent a novel therapeutic strategy for neurovascular protection in stroke.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Cisteína Endopeptidasas/deficiencia , Neuronas/metabolismo , Pericitos/metabolismo , Animales , Barrera Hematoencefálica/patología , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Técnicas de Cocultivo , Cisteína Endopeptidasas/genética , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología
8.
Mol Brain ; 13(1): 59, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32290845

RESUMEN

Sentrin/SUMO-specific protease 2 (SENP2) is a member of SENPs family involved in maturation of SUMO precursors and deSUMOylation of specific target, and is highly expressed in the central nervous system (CNS). Although SENP2 has been shown to modulate embryonic development, fatty acid metabolism, atherosclerosis and epilepsy, the function of SENP2 in the CNS remains poorly understood. To address the role of SENP2 in the CNS and its potential involvement in neuropathology, we generated SENP2 conditional knockout mice by crossing floxed SENP2 mice with CaMKIIα-Cre transgenic mice. Behavioral tests revealed that SENP2 ablation induced hyper-locomotor activity, anxiolytic-like behaviors, spatial working memory impairment and fear-associated learning defect. In line with these observations, our RNA sequencing (RNA-seq) data identified a variety of differential expression genes that are particularly enriched in locomotion, learning and memory related biologic process. Taken together, our results indicated that SENP2 plays a critical role in emotional and cognitive regulation. This SENP2 conditional knockout mice model may help reveal novel mechanisms that underlie a variety of neuropsychiatric disorders associated with anxiety and cognition.


Asunto(s)
Ansiedad/fisiopatología , Conducta Animal , Cisteína Endopeptidasas/deficiencia , Aprendizaje , Memoria , Neuronas/metabolismo , Prosencéfalo/metabolismo , Prosencéfalo/fisiopatología , Animales , Ansiedad/genética , Ansiedad/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Miedo , Regulación de la Expresión Génica , Ontología de Genes , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Circulation ; 141(23): 1885-1902, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32160764

RESUMEN

BACKGROUND: Immune checkpoint inhibitor (ICI) therapy is often accompanied by immune-related pathology, with an increasing occurrence of high-risk ICI-related myocarditis. Understanding the mechanisms involved in this side effect could enable the development of management strategies. In mouse models, immune checkpoints, such as PD-1 (programmed cell death protein 1), control the threshold of self-antigen responses directed against cardiac TnI (troponin I). We aimed to identify how the immunoproteasome, the main proteolytic machinery in immune cells harboring 3 distinct protease activities in the LMP2 (low-molecular-weight protein 2), LMP7 (low-molecular-weight protein 7), and MECL1 (multicatalytic endopeptidase complex subunit 1) subunit, affects TnI-directed autoimmune pathology of the heart. METHODS: TnI-directed autoimmune myocarditis (TnI-AM), a CD4+ T-cell-mediated disease, was induced in mice lacking all 3 immunoproteasome subunits (triple-ip-/-) or lacking either the gene encoding LMP2 and LMP7 by immunization with a cardiac TnI peptide. Alternatively, before induction of TnI-AM or after establishment of autoimmune myocarditis, mice were treated with the immunoproteasome inhibitor ONX 0914. Immune parameters defining heart-specific autoimmunity were investigated in experimental TnI-AM and in 2 cases of ICI-related myocarditis. RESULTS: All immunoproteasome-deficient strains showed mitigated autoimmune-related cardiac pathology with less inflammation, lower proinflammatory and chemotactic cytokines, less interleukin-17 production, and reduced fibrosis formation. Protection from TnI-directed autoimmune heart pathology with improved cardiac function in LMP7-/- mice involved a changed balance between effector and regulatory CD4+ T cells in the spleen, with CD4+ T cells from LMP7-/- mice showing a higher expression of inhibitory PD-1 molecules. Blocked immunoproteasome proteolysis, by treatment of TLR2 (Toll-like receptor 2)-engaged and TLR7 (Toll-like receptor 7)/TLR8 (Toll-like receptor 8)-engaged CD14+ monocytes with ONX 0914, diminished proinflammatory cytokine responses, thereby reducing the boost for the expansion of self-reactive CD4+ T cells. Correspondingly, in mice, ONX 0914 treatment reversed cardiac autoimmune pathology, preventing the induction and progression of TnI-AM when self-reactive CD4+ T cells were primed. The autoimmune signature during experimental TnI-AM, with high immunoproteasome expression, immunoglobulin G deposition, interleukin-17 production in heart tissue, and TnI-directed humoral autoimmune responses, was also present in 2 cases of ICI-related myocarditis, demonstrating the activation of heart-specific autoimmune reactions by ICI therapy. CONCLUSIONS: By reversing heart-specific autoimmune responses, immunoproteasome inhibitors applied to a mouse model demonstrate their potential to aid in the management of autoimmune myocarditis in humans, possibly including patients with ICI-related heart-specific autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Modelos Animales de Enfermedad , Eliminación de Gen , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunidad/inmunología , Miocarditis/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Anciano , Secuencia de Aminoácidos , Animales , Enfermedades Autoinmunes/inducido químicamente , Enfermedades Autoinmunes/genética , Cisteína Endopeptidasas/deficiencia , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/inmunología , Femenino , Humanos , Inmunidad/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Miocarditis/inducido químicamente , Miocarditis/genética , Complejo de la Endopetidasa Proteasomal/deficiencia , Complejo de la Endopetidasa Proteasomal/genética
10.
Acta Neuropathol Commun ; 7(1): 177, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31722749

RESUMEN

Intraneuronal insoluble inclusions made of Tau protein are neuropathological hallmarks of Alzheimer Disease (AD). Cleavage of Tau by legumain (LGMN) has been proposed to be crucial for aggregation of Tau into fibrils. However, it remains unclear if LGMN-cleaved Tau fragments accumulate in AD Tau inclusions.Using an in vitro enzymatic assay and non-targeted mass spectrometry, we identified four putative LGMN cleavage sites at Tau residues N167-, N255-, N296- and N368. Cleavage at N368 generates variously sized N368-Tau fragments that are aggregation prone in the Thioflavin T assay in vitro. N368-cleaved Tau is not detected in the brain of legumain knockout mice, indicating that LGMN is required for Tau cleavage in the mouse brain in vivo. Using a targeted mass spectrometry method in combination with tissue fractionation and biochemical analysis, we investigated whether N368-cleaved Tau is differentially produced and aggregated in brain of AD patients and control subjects. In brain soluble extracts, despite reduced uncleaved Tau in AD, levels of N368-cleaved Tau are comparable in AD and control hippocampus, suggesting that LGMN-mediated cleavage of Tau is not altered in AD. Consistently, levels of activated, cleaved LGMN are also similar in AD and control brain extracts. To assess the potential accumulation of N368-cleaved Tau in insoluble Tau aggregates, we analyzed sarkosyl-insoluble extracts from AD and control hippocampus. Both N368-cleaved Tau and uncleaved Tau were significantly increased in AD as a consequence of pathological Tau inclusions accumulation. However, the amount of N368-cleaved Tau represented only a very minor component (< 0.1%) of insoluble Tau.Our data indicate that LGMN physiologically cleaves Tau in the mouse and human brain generating N368-cleaved Tau fragments, which remain largely soluble and are present only in low proportion in Tau insoluble aggregates compared to uncleaved Tau. This suggests that LGMN-cleaved Tau has limited role in the progressive accumulation of Tau inclusions in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Cisteína Endopeptidasas/metabolismo , Agregado de Proteínas/fisiología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Animales , Encéfalo/patología , Cisteína Endopeptidasas/deficiencia , Cisteína Endopeptidasas/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Proteínas tau/genética
11.
Cell Rep ; 28(8): 2004-2011.e4, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31433978

RESUMEN

Brown adipose tissue (BAT) is a thermogenic organ that maintains body temperature and energy homeostasis. Transcriptional regulation plays an important role in the program of brown adipogenesis. However, it remains unclear how the transcriptional events are controlled in this program. In this study, we analyze an SENP2 BAT conditional knockout mouse model and find that SENP2-mediated de-SUMOylation is essential for BAT development. SENP2 catalyzes de-SUMOylation of cAMP response element-binding protein (CREB) to suppress Necdin expression, which induces brown adipocyte differentiation and brown adipogenesis. Mechanistically, we find that SUMOylation enhances CREB interaction with serine/threonine protein phosphatase 2A (PP2A) to de-phosphorylate CREB, which activates Necdin transcription. SENP2 deficiency enhances the expression of Necdin to inhibit brown adipocyte differentiation. Therefore, we reveal a crucial role of SENP2-mediated de-SUMOylation of CREB in suppression of Necdin expression during brown adipose development and brown adipogenesis.


Asunto(s)
Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Diferenciación Celular , Cisteína Endopeptidasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/crecimiento & desarrollo , Animales , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Cisteína Endopeptidasas/deficiencia , Humanos , Masculino , Ratones , Sumoilación
12.
Respir Med ; 155: 1-5, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31255902

RESUMEN

INTRODUCTION: Bleomycin pulmonary toxicity (BPT) is a potentially life-threatening consequence of bleomycin usage in patients. An overproduction of epithelium-derived cytokines, habitually linked to allergic inflammation, has been recently revealed in experimental models of BPT. METHODS: We assessed retrospectively our cohort of patients with Hodgkin Lymphoma treated with bleomycin between 2014 and 2016 for their demographic, clinical features, including BPT development, atopy status and risk factors for BPT. Then they were invited for allergy testing and blood sample collection. The samples were stimulated with different stimuli (Bleomycin, IL-33, TSLP) for 24 h on cell culture. The culture supernatants were analysed for TGF-ß, Galectin3, Arginin, Amphiregulin, Eotaxin, IFNγ, TNFα, IL1ß, 4, 5, 6, 10, 13, 17, MIP-1α, and bleomycin hydrolase (BLH) levels. RESULTS: The cohort consisted of 51 patients showed that atopy was the only significant risk factor for BPT occurrence (OR: 7.2, p = 0.007). Fourteen subjects were included for blood analysis. The analysis of supernatants at the unstimulated condition revealed that BLH and Amphiregulin were significantly lower in patients who had BPT than controls. The BLH cut-off that best identified a history of BPT was 175.31 (Sensitivity: 62.5%, specificity: 100%). Following the stimulation, BLH reduced compared to the unstimulated condition and the difference between groups remained significant (p < 0.05). CONCLUSION: Our study is the first to report that low levels of bleomycin hydrolase in allergic individuals may be predisposing to a possible pathway of fibrosis.


Asunto(s)
Bleomicina/toxicidad , Hipersensibilidad Inmediata , Adulto , Anfirregulina , Estudios de Cohortes , Cisteína Endopeptidasas/deficiencia , Femenino , Enfermedad de Hodgkin/tratamiento farmacológico , Humanos , Hipersensibilidad/enzimología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo , Adulto Joven
13.
Nat Immunol ; 20(7): 879-889, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31182807

RESUMEN

CD8+ T cells and natural killer (NK) cells are central cellular components of immune responses against pathogens and cancer, which rely on interleukin (IL)-15 for homeostasis. Here we show that IL-15 also mediates homeostatic priming of CD8+ T cells for antigen-stimulated activation, which is controlled by a deubiquitinase, Otub1. IL-15 mediates membrane recruitment of Otub1, which inhibits ubiquitin-dependent activation of AKT, a kinase that is pivotal for T cell activation and metabolism. Otub1 deficiency in mice causes aberrant responses of CD8+ T cells to IL-15, rendering naive CD8+ T cells hypersensitive to antigen stimulation characterized by enhanced metabolic reprograming and effector functions. Otub1 also controls the maturation and activation of NK cells. Deletion of Otub1 profoundly enhances anticancer immunity by unleashing the activity of CD8+ T cells and NK cells. These findings suggest that Otub1 controls the activation of CD8+ T cells and NK cells by functioning as a checkpoint of IL-15-mediated priming.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Cisteína Endopeptidasas/metabolismo , Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Cisteína Endopeptidasas/deficiencia , Enzimas Desubicuitinizantes/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucina-15/genética , Melanoma Experimental , Ratones , Ratones Transgénicos , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Interleucina-15/metabolismo , Autotolerancia/genética , Autotolerancia/inmunología , Transducción de Señal , Especificidad del Receptor de Antígeno de Linfocitos T , Ubiquitinación
14.
Cancer Res ; 79(15): 3891-3902, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31186231

RESUMEN

Myeloid-derived suppressor cells (MDSC) can suppress immunity and promote tumorigenesis, and their abundance is associated with poor prognosis. In this study, we show that SUMO1/sentrin-specific peptidase 1 (SENP1) regulates the development and function of MDSC. SENP1 deficiency in myeloid cells promoted MDSC expansion in bone marrow, spleen, and other organs. Senp1-/- MDSC showed stronger immunosuppressive activity than Senp1+/+ MDSC; we observed no defects in the differentiation of myeloid precursor cell in Senp1-/- mice. Mechanistically, SENP1-mediated regulation of MDSC was dependent on STAT3 signaling. We identified CD45 as a specific STAT3 phosphatase in MDSC. CD45 was SUMOylated in MDSC and SENP1 could deconjugate SUMOylated CD45. In Senp1-/- MDSC, CD45 was highly SUMOylated, which reduced its phosphatase activity toward STAT3, leading to STAT3-mediated MDSC development and function. These results reveal a suppressive function of SENP1 in modulating MDSC expansion and function via CD45-STAT3 signaling axis. SIGNIFICANCE: These findings show that increased SUMOylation of CD45 via loss of SENP1 suppresses CD45-mediated dephosphorylation of STAT3, which promotes MDSC development and function, leading to tumorigenesis.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Animales , Carcinogénesis , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Cisteína Endopeptidasas/biosíntesis , Cisteína Endopeptidasas/deficiencia , Femenino , Células HEK293 , Humanos , Antígenos Comunes de Leucocito/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Noqueados , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/patología , Factor de Transcripción STAT3/metabolismo , Sumoilación
15.
Cell Res ; 29(6): 474-485, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31086255

RESUMEN

NF-κB, a family of transcription factors regulating diverse biological processes including immune responses, is activated by canonical and noncanonical pathways based on degradation of IκBα and processing of the IκB-like protein p100, respectively. Although p100 responds to noncanonical NF-κB stimuli for processing, it does not undergo degradation, but rather becomes accumulated, along with canonical NF-κB activation. We show here that the stability of p100 is tightly controlled by a deubiquitinase, Otub1. Otub1 deficiency not only promotes signal-induced p100 processing and noncanonical NF-κB activation but also causes steady-state p100 degradation, leading to aberrant NF-κB activation in the canonical pathway. B-cell-conditional deletion of Otub1 results in B-cell hyperplasia, antibody hyper-production, and lupus-like autoimmunity. Otub1-deficient B cells display aberrantly activated phenotypes and overproduce the cytokine IL-6, contributing to autoimmunity induction. Thus, maintenance of p100 stability by Otub1 serves as an unusual mechanism of NF-κB regulation that prevents autoimmunity.


Asunto(s)
Autoinmunidad , Cisteína Endopeptidasas/metabolismo , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Animales , Células Cultivadas , Cisteína Endopeptidasas/deficiencia , Enzimas Desubicuitinizantes , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Estabilidad Proteica
16.
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
17.
Brain Res Bull ; 142: 147-155, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30030107

RESUMEN

Mammalian asparagine endopeptidase (AEP) is a lysosomal cysteine protease that cleaves protein substrates on the C-terminal side of asparagine. The expression and activity of AEP are closely related to many pathological conditions that include cancer, atherosclerosis and inflammation. It has been validated that the level of AEP is elevated in aged human and neurodegenerative diseases like Alzheimer's disease (AD). Mood disorder is one of the most emotional symptoms that can be seen in AD patients, which leads us to assume that AEP can modulate affective behaviors. AEP knockout (AEP KO) and wildtype (WT) mice were used in this study, and a series of behavioral tests were performed to establish a potential link between AEP and psychiatric disorders. It was demonstrated that AEP KO mice displayed lower anxiety-like behavior and more advance exploratory behavior in open-field and hole-board tests. AEP KO mice reduced depressive-like behaviors in the forced swim and tail suspension tests. Morris water maze (MWM) test showed that the abilities of spatial learning and memory were elevated in AEP-deletion mice compared with those of WT mice. Furthermore, the enhanced synaptic plasticity (LTP and DPT) as well as the increased expressions of SYP and PSD-95 proteins in hippocampus were showed in AEP KO mice. Otherwise, the level of BDNF protein was reduced and the level of NF-κB p65 protein was increased in hippocampus and frontal cortex of AEP KO mice. These data highlight the importance of studying AEP in the anxiety and depression behaviors and the spatial learning and memory.


Asunto(s)
Ansiedad/enzimología , Cisteína Endopeptidasas/deficiencia , Depresión/enzimología , Conducta Exploratoria/fisiología , Aprendizaje por Laberinto/fisiología , Memoria Espacial/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cognición/fisiología , Cisteína Endopeptidasas/genética , Homólogo 4 de la Proteína Discs Large/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Lóbulo Frontal/enzimología , Hipocampo/enzimología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Plasticidad Neuronal/fisiología , Sinaptofisina/metabolismo
18.
Immunol Cell Biol ; 96(9): 981-993, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29738610

RESUMEN

The role of the immunoproteasome is perceived as confined to adaptive immune responses given its ability to produce peptides ideal for MHC Class-I binding. Here, we demonstrate that the immunoproteasome subunit, LMP2, has functions beyond its immunomodulatory role. Using LMP2-deficient mice, we demonstrate that LMP2 is crucial for lymphocyte development and survival in the periphery. Moreover, LMP2-deficient lymphocytes show impaired degradation of key BH3-only proteins, resulting in elevated levels of pro-apoptotic BIM and increased cell death. Interestingly, LMP2 is the sole immunoproteasome subunit required for BIM degradation. Together, our results suggest LMP2 has important housekeeping functions and represents a viable therapeutic target for cancer.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/inmunología , Cisteína Endopeptidasas/inmunología , Linfocitos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Animales , Western Blotting , Línea Celular , Supervivencia Celular , Células Cultivadas , Cisteína Endopeptidasas/deficiencia , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/deficiencia
19.
Cell Death Dis ; 8(8): e2970, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28771229

RESUMEN

In the last years, autophagy has been revealed as an essential pathway for multiple biological processes and physiological functions. As a catabolic route, autophagy regulation by nutrient availability has been evolutionarily conserved from yeast to mammals. On one hand, autophagy induction by starvation is associated with a significant loss in body weight in mice. Here, we demonstrate that both genetic and pharmacological inhibition of the autophagy process compromise weight loss induced by starvation. Moreover, autophagic potential also impacts on weight gain induced by distinct hypercaloric regimens. Atg4b-deficient mice, which show limited autophagic competence, exhibit a major increase in body weight in response to distinct obesity-associated metabolic challenges. This response is characterized by the presence of larger adipocytes in visceral fat tissue, increased hepatic steatosis, as well as reduced glucose tolerance and attenuated insulin responses. Similarly, autophagy-deficient mice are more vulnerable to experimentally induced type-I diabetes, showing an increased susceptibility to acute streptozotocin administration. Notably, pharmacological stimulation of autophagy in wild-type mice by spermidine reduced both weight gain and obesity-associated alterations upon hypercaloric regimens. Altogether, these results indicate that systemic autophagic activity influences the resilience of the organism to weight gain induced by high-calorie diets, as well as to the obesity-associated features of both type-1 and type-2 diabetes.


Asunto(s)
Autofagia , Dieta/efectos adversos , Células Secretoras de Insulina/metabolismo , Obesidad/inducido químicamente , Obesidad/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Animales , Proteínas Relacionadas con la Autofagia/deficiencia , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Cisteína Endopeptidasas/deficiencia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Hígado Graso/inducido químicamente , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Células Secretoras de Insulina/patología , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/patología
20.
Arterioscler Thromb Vasc Biol ; 37(9): 1698-1709, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28751569

RESUMEN

OBJECTIVE: Transdifferentiation of adventitial fibroblasts (AFs) into myofibroblasts plays a critical role during the vascular remodeling that occurs during atherosclerosis, restenosis, and aortic aneurysm. The ubiquitination/deubiquitination regulatory system is essential for the quality control of proteins. The involvement of ubiquitination/deubiquitination during AF transdifferentiation remains largely unknown. In this study, we determined the role of cylindromatosis (CYLD), a deubiquitinase, in the process of AF differentiation and activation in vitro and in vivo. APPROACH AND RESULTS: Transforming growth factor-ß1 and homocysteine, 2 known inducers of AF transdifferentiation, greatly upregulated CYLD expression in a time- and dose-dependent manner. The silencing of CYLD significantly inhibited AF transdifferentiation and activation as evidenced by the expression of contractile proteins, the production of the proinflammatory cytokines MCP-1 (monocyte chemotactic protein 1) and IL-6 (interleukin-6), the deposition of extracellular matrix, and cell migration. We further asked whether CYLD mediates AF activation via the regulation of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) as it is an essential factor during AF transdifferentiation. Indeed, the silencing of CYLD repressed transforming growth factor-ß1-induced and homocysteine-induced Nox4 upregulation and reactive oxygen species production, whereas Nox4 overexpression greatly rescued the inhibitory effect on AF activation by CYLD silencing. Most interestingly, transforming growth factor-ß1 and homocysteine repressed Nox4 ubiquitination and prolonged the half-life of Nox4. Moreover, Nox4 was deubiquitinated via a direct interaction with the ubiquitin-specific protease domain of CYLD. In accordance, hyperhomocysteinemia significantly increased adventitial CYLD and Nox4 expression, promoted AF transdifferentiation, and aggravated CaPO4-induced abdominal aortic aneurysm in mice. These effects were abolished in CYLD-/- mice. CONCLUSIONS: CYLD contributes to the transdifferentiation of AFs via deubiquitinating Nox4 and may play a role in vascular remodeling.


Asunto(s)
Adventicia/enzimología , Aneurisma de la Aorta Abdominal/enzimología , Transdiferenciación Celular , Cisteína Endopeptidasas/metabolismo , Miofibroblastos/enzimología , NADPH Oxidasas/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Remodelación Vascular , Adventicia/efectos de los fármacos , Adventicia/patología , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Células COS , Fosfatos de Calcio , Movimiento Celular , Transdiferenciación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Chlorocebus aethiops , Cisteína Endopeptidasas/deficiencia , Cisteína Endopeptidasas/genética , Enzima Desubiquitinante CYLD , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estabilidad de Enzimas , Matriz Extracelular/metabolismo , Genotipo , Células HEK293 , Semivida , Homocisteína/farmacología , Humanos , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/enzimología , Hiperhomocisteinemia/genética , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Fenotipo , Proteolisis , Interferencia de ARN , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta1/farmacología , Ubiquitina Tiolesterasa/genética , Ubiquitinación , Remodelación Vascular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...