Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nat Immunol ; 21(10): 1205-1218, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32839608

RESUMO

Immune-modulating therapies have revolutionized the treatment of chronic diseases, particularly cancer. However, their success is restricted and there is a need to identify new therapeutic targets. Here, we show that natural killer cell granule protein 7 (NKG7) is a regulator of lymphocyte granule exocytosis and downstream inflammation in a broad range of diseases. NKG7 expressed by CD4+ and CD8+ T cells played key roles in promoting inflammation during visceral leishmaniasis and malaria-two important parasitic diseases. Additionally, NKG7 expressed by natural killer cells was critical for controlling cancer initiation, growth and metastasis. NKG7 function in natural killer and CD8+ T cells was linked with their ability to regulate the translocation of CD107a to the cell surface and kill cellular targets, while NKG7 also had a major impact on CD4+ T cell activation following infection. Thus, we report a novel therapeutic target expressed on a range of immune cells with functions in different immune responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Inflamação/imunologia , Células Matadoras Naturais/imunologia , Leishmania donovani/fisiologia , Leishmaniose Visceral/imunologia , Malária/imunologia , Proteínas de Membrana/metabolismo , Plasmodium/fisiologia , Animais , Células Cultivadas , Citotoxicidade Imunológica , Modelos Animais de Doenças , Exocitose , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Vesículas Secretórias/metabolismo
3.
Hepatology ; 2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37820061

RESUMO

BACKGROUND AND AIMS: Cancer stem cells (CSCs) contribute to therapy resistance in HCC. Linear ubiquitin chain assembly complex (LUBAC) has been reported to accelerate the progression of cancers, yet its role in the sorafenib response of HCC is poorly defined. Herein, we investigated the impact of LUBAC on sorafenib resistance and the CSC properties of HCC, and explored the potential targeted drugs. APPROACH AND RESULTS: We found that HOIL-1, but not the other components of LUBAC, played a contributing role in LUBAC-mediated HCC sorafenib resistance, independent of its ubiquitin ligase activity. Both in vitro and in vivo assays revealed that the upregulated HOIL-1 expression enhanced the CSC properties of HCC. Mechanistically, HOIL-1 promoted sorafenib resistance and the CSC properties of HCC through Notch1 signaling. Mass spectrometry, co-immunoprecipitation, western blot, and immunofluorescence were used to determine that the A64/Q65 residues of HOIL-1 bound with the K78 residue of Numb, resulting in impaired Numb-mediated Notch1 lysosomal degradation. Notably, pixantrone was screened out by Autodock Vina, which was validated to disrupt HOIL-1/Numb interaction to inhibit Notch1 signaling and CSC properties by targeting the Q65 residue of HOIL-1. Moreover, pixantrone exerted synergistic effects with sorafenib for the treatment of HCC in different HCC mouse models. CONCLUSIONS: HOIL-1 is critical in promoting sorafenib resistance and CSC properties of HCC through Notch1 signaling. Pixantrone targeting HOIL-1 restrains the sorafenib resistance and provides a potential therapeutic intervention for HCC.

4.
Acta Pharmacol Sin ; 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38740904

RESUMO

The circadian clock is the inner rhythm of life activities and is controlled by a self-sustained and endogenous molecular clock, which maintains a ~ 24 h internal oscillation. As the core element of the circadian clock, BMAL1 is susceptible to degradation through the ubiquitin-proteasome system (UPS). Nevertheless, scant information is available regarding the UPS enzymes that intricately modulate both the stability and transcriptional activity of BMAL1, affecting the cellular circadian rhythm. In this work, we identify and validate UBR5 as a new E3 ubiquitin ligase that interacts with BMAL1 by using affinity purification, mass spectrometry, and biochemical experiments. UBR5 overexpression induced BMAL1 ubiquitination, leading to diminished stability and reduced protein level of BMAL1, thereby attenuating its transcriptional activity. Consistent with this, UBR5 knockdown increases the BMAL1 protein. Domain mapping discloses that the C-terminus of BMAL1 interacts with the N-terminal domains of UBR5. Similarly, cell-line-based experiments discover that HYD, the UBR5 homolog in Drosophila, could interact with and downregulate CYCLE, the BMAL1 homolog in Drosophila. PER2-luciferase bioluminescence real-time reporting assay in a mammalian cell line and behavioral experiments in Drosophila reveal that UBR5 or hyd knockdown significantly reduces the period of the circadian clock. Therefore, our work discovers a new ubiquitin ligase UBR5 that regulates BMAL1 stability and circadian rhythm and elucidates the underlying molecular mechanism. This work provides an additional layer of complexity to the regulatory network of the circadian clock at the post-translational modification level, offering potential insights into the modulation of the dysregulated circadian rhythm.

5.
J Autoimmun ; 135: 102993, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36642058

RESUMO

BACKGROUND & AIMS: The N6-methyladenosine (m6A) reader YTH domain-containing family protein 2 (YTHDF2) is critically involved in a multiplicity of biological processes by mediating the degradation of m6A modified mRNAs. Based on our current understanding of this process, we hypothesized that YTHDF2 will play a role in the natural history and function of myeloid-derived suppressor cells (MDSC) and in particular in AIH. APPROACH & RESULTS: We took advantage of YTHDF2 conditional knock-out mice to first address the phenotype and function of MDSCs by flow cytometry. Importantly, the loss of YTHDF2 resulted in a gradual elevation of MDSCs including PMN-MDSCs both in liver and ultimately in the BM. Notably, YTHDF2 deficiency in myeloid cells attenuated concanavalin (ConA)-induced liver injury, with enhanced expansion and chemotaxis to liver. Furthermore, MDSCs from Ythdf2CKO mice had a greater suppressive ability to inhibit the proliferation of T cells. Using multi-omic analysis of m6A RNA immunoprecipitation (RIP) and mRNA sequencing, we noted RXRα as potential target of YTHDF2. Indeed YTHDF2-RIP-qPCR confirmed that YTHDF2 directly binds RXRα mRNA thus promoting degradation and decreasing gene expression. Finally, by IHC and immunofluorescence, YTHDF2 expression was significantly upregulated in the liver of patients with AIH which correlated with the degree of inflammation. CONCLUSION: Suppression of YTHDF2 enhances the expansion, chemotaxis and suppressive function of MDSCs and our data reveals a unique therapeutical target in immune mediated hepatitis.


Assuntos
Hepatite Autoimune , Células Supressoras Mieloides , Animais , Camundongos , Células Mieloides , Linfócitos T , Fatores de Transcrição/metabolismo
6.
Ecotoxicol Environ Saf ; 263: 115366, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37573610

RESUMO

Advanced oxidation processes (AOPs) based on ultrasound (US) have attracted considerable attention in recent years due to its advantages in the degradation of landfill leachate. The review summarizes the existing treatment methods of leachate from lab-scale, compares their advantages and disadvantages by focusing on the degradation of emerging contaminants (ECs) in the leachate. Then the US-based AOPs are introduced emphatically, including their degradation mechanisms, influencing factors, energy consumption, further optimization methods as well as the possibility of field-scale application are systematically described. Moreover, this review also expounds on the advantages of dual-frequency US (DFUS) technology compared with single-frequency US, and a theoretically feasible DFUS process is proposed to treat ECs in the leachate. Finally, suggestions and prospects for US technologies in treating landfill leachate are put forward to aid future research on landfill leachate treatment. Meaningfully, this manuscript will provide reference values of US-based technologies in landfill leachate treatment for the practical use, facilitating the development of US-based AOPs in landfill leachate management and disposal.


Assuntos
Poluentes Químicos da Água , Oxirredução
7.
J Am Chem Soc ; 144(48): 22202-22211, 2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36404600

RESUMO

The absolute coverage of CO has been a missing piece in the mechanistic puzzle of the CO reduction reaction (CORR) on Cu. For the first time, we revealed the upper bound of the CO coverage under electrocatalytic conditions to be 0.05 monolayer at atmospheric pressure and the saturation CO coverage to be ∼0.25 monolayer by conducting surface enhanced infrared spectroscopy at CO pressures up to 60 barg in a custom-designed spectroelectrochemical cell. CORR activities on Cu were also determined in the same pressure range. Calculated reaction orders of C2+ products with respect to adsorbed CO are substantially less than unity, clearly indicating that the coupling of adsorbed CO is not the rate-determining step leading to multicarbon products. The increase in CO coverage can reduce the C affinity on the Cu surface and favor the selectivity towards oxygenates, especially acetate, over ethylene. Uncommon products, including ethane, glycolaldehyde, and ethylene glycol, were detected in appreciable amounts, likely due to a new C-C coupling mechanism taking place at elevated CO pressures.


Assuntos
Etano , Espectrofotometria Infravermelho
8.
J Am Chem Soc ; 144(44): 20495-20506, 2022 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-36286405

RESUMO

Understanding of the reaction network of Cu-catalyzed CO2/CO electroreduction reaction [CO(2)RR] remains incomplete despite intense research efforts. This is in part because the rate-determining step occurs early in the reaction network, leading to short lifetimes of subsequent surface-bound intermediates, the knowledge of which is key to selectivity control. In this work, we demonstrate that alkyl groups can effectively couple with surface intermediates in the Cu-catalyzed CORR and, for the first time, intercept elusive C1 and C2 intermediates. Combined reactivity data and in situ spectroscopic results demonstrated that surface-bound alkyl groups derived from the corresponding alkyl iodides are able to couple with adsorbed CO to form carboxylates and ketones via one and two successive nucleophilic attacks, respectively. Leveraging this new chemistry, CHx (x ≤ 3) and C2Hx (x ≤ 4) are intercepted and identified as precursors for methane and n-propanol in the CORR, respectively. Importantly, reaction pathways leading to methane and C2+ products are not intrinsically orthogonal, but their connection is mainly impeded by low coverages of energetic intermediates. This study shows that perturbing the reaction of interest by introducing a slightly interacting probe reaction network could be an effective and general strategy in mechanistic studies of catalytic reactions.

9.
10.
J Hepatol ; 72(1): 167-182, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31449859

RESUMO

Basic and clinical studies have demonstrated the efficacy of immunotherapy, a technical and conceptual breakthrough that has revolutionised cancer treatment. Hepatocellular carcinoma (HCC), a deadly malignancy with aetiologic diversity and a chronic course, is strongly influenced by the immune system, and was recently found to partially benefit from immune-checkpoint inhibitor therapy. Notably, HCC onco-immunology depends on diverse genetic and environmental factors that together shape cancer-promoting inflammation and immune dysfunction - critical processes that control HCC malignant progression and response to therapy. Herein, we summarise the current understanding of liver and HCC onco-immunology obtained through basic studies with mouse models and clinical practice in humans. In particular, we discuss preclinical and clinical findings that implicate immunomodulation as a major factor in HCC development and explain the basis for HCC-targeting immunotherapy.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/terapia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia Adotiva/métodos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/terapia , Microambiente Tumoral/imunologia , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Humanos , Fígado/imunologia , Fígado/patologia , Camundongos
11.
Mol Cancer ; 18(1): 163, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31735169

RESUMO

BACKGROUND: Dynamic N6-methyladenosine (m6A) modification was previously identified as a ubiquitous post-transcriptional regulation that affected mRNA homeostasis. However, the m6A-related epitranscriptomic alterations and functions remain elusive in human cancer. Here we aim to identify the profile and outcome of m6A-methylation in hepatocellular carcinoma (HCC). RESULTS: Using liquid chromatography-tandem mass spectrometry and m6A-immunoprecipitation in combination with high-throughput sequencing, we determined the m6A-mRNA levels in human HCC. Human HCC exhibited a characteristic gain of m6A modification in tandem with an increase of mRNA expression, owing to YTH domain family 2 (YTHDF2) reduction. The latter predicted poor classification and prognosis of HCC patients, and highly correlated with HCC m6A landscape. YTHDF2 silenced in human HCC cells or ablated in mouse hepatocytes provoked inflammation, vascular reconstruction and metastatic progression. Mechanistically, YTHDF2 processed the decay of m6A-containing interleukin 11 (IL11) and serpin family E member 2 (SERPINE2) mRNAs, which were responsible for the inflammation-mediated malignancy and disruption of vascular normalization. Reciprocally, YTHDF2 transcription succumbed to hypoxia-inducible factor-2α (HIF-2α). Administration of a HIF-2α antagonist (PT2385) restored YTHDF2-programed epigenetic machinery and repressed liver cancer. CONCLUSION: Our results have characterized the m6A-mRNA landscape in human HCC and revealed YTHDF2 as a molecular 'rheostat' in epitranscriptome and cancer progression.


Assuntos
Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/patologia , Inflamação/complicações , Inflamação/genética , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/patologia , Neovascularização Patológica/genética , Proteínas de Ligação a RNA/genética , Adenosina/análogos & derivados , Animais , Epigênese Genética , Humanos , Metilação , Camundongos , Modelos Biológicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transcriptoma
13.
J Immunol ; 193(10): 5161-70, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25339662

RESUMO

Previous investigations demonstrated that protein tyrosine phosphatase, receptor type, O (PTPRO) acts as a tumor suppressor in liver cancer; however, little is known about its role in liver inflammation. Thus, we investigated the role of PTPRO in fulminant hepatitis (FH) using a Con A-induced mouse model. Significantly more severe liver damage, but attenuated inflammation, was detected in PTPRO-knockout (KO) mice, and PTPRO deficiency could confer this phenotype to wild-type mice in bone marrow transplantation. Moreover, hepatocytes with PTPRO depletion were more sensitive to TNF-α-induced apoptosis, and secretion of cytokines was significantly decreased in both T and NK/NKT cells and led to marked impairment of NF-κB activation. Intriguingly, wild-type and PTPRO-KO cells responded equally to TNF-α in activation of IKK, but NF-κB activation was clearly decreased in PTPRO-KO cells. PTPRO associated with ErbB2, and loss of PTPRO potentiated activation of the ErbB2/Akt/GSK-3ß/ß-catenin cascade. Increased ß-catenin formed a complex with NF-κB and attenuated its nuclear translocation and activation. Importantly, in humans, PTPRO was much decreased in FH, and this was associated with enhanced ß-catenin accumulation but reduced IFN-γ secretion. Taken together, our study identified a novel PTPRO/ErbB2/Akt/GSK-3ß/ß-catenin/NF-κB axis in FH, which suggests that PTPRO may have therapeutic potential in this liver disease.


Assuntos
Hepatite Animal/imunologia , Hepatócitos/imunologia , Fígado/imunologia , NF-kappa B/imunologia , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/imunologia , beta Catenina/imunologia , Doença Aguda , Animais , Concanavalina A , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/imunologia , Glicogênio Sintase Quinase 3 beta , Hepatite Animal/induzido quimicamente , Hepatite Animal/mortalidade , Hepatite Animal/patologia , Hepatócitos/patologia , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/mortalidade , Inflamação/patologia , Interferon gama/imunologia , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , NF-kappa B/agonistas , NF-kappa B/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/deficiência , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Índice de Gravidade de Doença , Transdução de Sinais , Análise de Sobrevida , Linfócitos T/imunologia , Linfócitos T/patologia , beta Catenina/genética
14.
Cell Physiol Biochem ; 37(1): 214-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26303994

RESUMO

BACKGROUND/AIMS: Critical roles of PTPRO and TLR4 have been implicated in hepatocellular carcinoma. However, little is known about their modifying effects on inflammation-related diseases in liver, particularly fulminant hepatitis (FH). We aim to investigate the potential role of PTPRO and its interaction with TLR4 in LPS/D-GaIN induced FH. METHODS: A LPS/D-GaIN induced mouse FH model was used. RAW264.7 cells were transfected with PTPRO over-expressed lentiviral plasmids for further investigation. RESULTS: The mortality of PTPRO KO mice is higher than WT mice after LPS/D-GaIN administration. Aggravated liver injury was demonstrated by increased level of serous ALT and AST and numerous hepatic cells death in PTPRO KO mice following LPS/D-GaIN administration. Interestingly, inflammation was attenuated in PTPRO-deficient mice following LPS/D-GaIN administration, which was suggested by decreased inflammatory cytokines (TNF-α, IFN-γ, IL-1ß, IL-6, IL-17A and IL-12) and cells infiltrating into spleen (CD3(+)IFN-γ(+) cells, CD3(+)TNF-α(+) cells, F4/80(+)/TLR4(+) cells). A feedback regulation between PTPRO and TLR4 dependent on NF-κB signaling pathway was demonstrated in vivo and in vitro. CONCLUSION: PTPRO plays an important role in FH by interacting with TLR4. The crosstalk between PTPRO and TLR4 is a novel bridge linking innate immune and adaptive immune in acute liver injury.


Assuntos
Galactosamina/farmacologia , Hepatite/metabolismo , Hepatite/patologia , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/deficiência , Imunidade Adaptativa/efeitos dos fármacos , Animais , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Imunidade Inata/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/patologia , Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Baço/efeitos dos fármacos , Baço/metabolismo , Baço/patologia , Receptor 4 Toll-Like/metabolismo
15.
Cell Physiol Biochem ; 35(3): 885-98, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25633279

RESUMO

BACKGROUND/AIMS: PTPRO (protein tyrosine phosphatase, receptor type O) is implicated in diverse physiological and pathological processes in cancer and hepatic ischemia/reperfusion injury, although little is known about its role in hepatic fibrosis. METHODS: Here, by using genetically deficient mice, we reported that PTPRO knockout (PTPRO(-/-)) significantly attenuated liver injury, release of inflammatory factors, tissue remodeling, and liver fibrosis in two experimental mouse models of fibrogenesis induced by bile-duct ligation or carbon tetrachloride administration. RESULTS: However, we proved that PTPRO expression was strongly downregulated in clinical and experimental liver fibrosis specimens. Further investigations revealed that stimulation of primary hepatic stellate cells (HSCs) and hepatocytes with specific activator platelet-derived growth factor (PDGF)-BB increased PTPRO transcription in HSCs but had the opposite effect in primary hepatocytes. More importantly, synthetic short hairpin RNA targeting PTPRO significantly neutralized PDGF-BB-induced HSC proliferation and myofibroblast marker expression through downregulated phosphorylation of extracellular signal-regulated kinase (ERK) and AKT. CONCLUSION: These observations confirm that PTPRO plays a critical role in liver fibrogenesis by affecting PDGF signaling in HSC activation and might be developed into a feasible therapeutic approach for the treatment of chronic fibrotic liver diseases.


Assuntos
Proliferação de Células/genética , Cirrose Hepática/genética , Proteínas Proto-Oncogênicas c-sis/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Animais , Becaplermina , Tetracloreto de Carbono/toxicidade , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Hepatócitos/efeitos dos fármacos , Humanos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-sis/administração & dosagem , RNA Interferente Pequeno , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/biossíntese , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Transdução de Sinais
16.
J Hepatol ; 60(2): 306-12, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24128416

RESUMO

BACKGROUND & AIMS: Nuclear factor-κB (NF-κB) activation in hepatocytes and macrophages appeared as a double-edged-sword in hepatic ischemia reperfusion (IR) injury. Protein tyrosine phosphatase receptor type O (PTPRO) was recently identified as a potential activator of c-Src, which can in turn activate the NF-κB pathway. In this study, we aimed to determine the change and function of PTPRO in hepatocytes and macrophages during IR. METHODS: Clinical patients with benign liver condition undergoing liver surgery were recruited in our study. Wild type (WT) and ptpro(-/-) C57BL/6 mice were processed to construct hepatic IR models. Isolated mouse hepatocytes and macrophages were treated with peroxide or TNFα in vitro. RESULTS: In human and mouse IR models, PTPRO level was decreased in the early phase but reversed in the late phase. In vitro studies demonstrated that NF-κB up-regulated PTPRO transcription. Using ptpro(-/-) mice and primary cells, we found that PTPRO deficiency resulted in reduction of NF-κB activation in both hepatocytes and macrophages and was correlated to c-Src phosphorylation; PTPRO in hepatocytes alleviated, but PTPROt in macrophages exacerbated IR injury. CONCLUSIONS: PTPRO activates NF-κB in a positive feedback manner, and plays a dual role in hepatic IR injury.


Assuntos
Fígado/enzimologia , Fígado/lesões , NF-kappa B/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Traumatismo por Reperfusão/enzimologia , Animais , Proteína Tirosina Quinase CSK , Modelos Animais de Doenças , Retroalimentação Fisiológica , Expressão Gênica , Hepatócitos/enzimologia , Humanos , Fígado/patologia , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/deficiência , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Quinases da Família src/metabolismo
17.
Hepatology ; 57(2): 678-88, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22821478

RESUMO

UNLABELLED: Protein tyrosine phosphatase receptor type O (PTPRO), one of the receptor types of phosphotyrosine phosphatases (PTP), was recently described as a tumor suppressor in various kinds of cancers. We aimed to clarify the role of PTPRO in hepatocellular carcinoma (HCC). It was demonstrated in 180 pairs (120 male and 60 female) of clinical HCC specimens that the PTPRO level was significantly reduced, as compared with adjacent tissue, and the PTPRO level in male adjacent tissue was lower than in female. We further found that estrogen receptor alpha (ERα) could up-regulate PTPRO expression as a transcription factor. Moreover, an in vitro study showed that cell proliferation was inhibited and apoptosis was promoted in PTPRO-transduced HCC cell lines, whereas an in vivo study represented that tumor number and size was increased in ptpro(-/-) mice. As a result of its tumor-suppressive position, PTPRO was proved to down-regulate signal transducers and activators of transcription (STAT3) activity dependent on Janus kinase 2 (JAK2) and phosphoinositide 3-kinase (PI3K) dephosphorylation. CONCLUSIONS: PTPRO expression results in pathological deficiency and gender bias in HCC, which could be attributed to ERα regulation. The suppressive role of PTPRO in HCC could be ascribed to STAT3 inactivation.


Assuntos
Carcinoma Hepatocelular/patologia , Receptor alfa de Estrogênio/fisiologia , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/fisiologia , Fator de Transcrição STAT3/genética , Animais , Apoptose , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Dietilnitrosamina , Progressão da Doença , Regulação para Baixo , Receptor alfa de Estrogênio/biossíntese , Feminino , Humanos , Janus Quinase 2/metabolismo , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Fosfatidilinositol 3-Quinases , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/biossíntese , Fatores Sexuais , Proteínas Supressoras de Tumor/fisiologia
18.
Tumour Biol ; 35(10): 10267-73, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25034527

RESUMO

Protein tyrosine phosphatase receptor type O (PTPRO) has been identified as a tumor suppressor in a number of cancers including hepatocellular carcinoma (HCC). Toll-like receptor 4 (TLR4) plays diverse roles in HCC tumorigenesis and progression. The association between PTPRO and TLR4 signaling in HCC remains largely unknown. We aimed to clarify the interaction between PTPRO and TLR4 in HCC. Surprisingly, we found reduced and positive-related expression of TLR4 and PTPRO in 84 human HCC specimens. Increased TLR4 expression and activity was found in PTPRO-overexpressed HCC cells stimulated with lipopolysaccharide (LPS). The feedback regulation of PTPRO and TLR4 was dependent on nuclear factor-κB (NF-κB) activation, as suggested by NF-κB inhibition and luciferase reporter assay. Our study suggests that the effect of PTPRO on TLR4 signaling is dependent on NF-κB pathway, suggesting an interesting PTPRO/TLR4/NF-κB signaling feedback loop in HCC carcinogenesis and progression.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo , Western Blotting , Linhagem Celular Tumoral , Imunofluorescência , Humanos , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transfecção
19.
Nat Commun ; 15(1): 1926, 2024 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-38431637

RESUMO

Understanding the structure of the electric double layer (EDL) is critical for designing efficient electrocatalytic processes. However, the interplay between reactant adsorbates and the concentrated ionic species within the EDL remains an aspect that has yet to be fully explored. In the present study, we employ electrochemical CO reduction on Cu as a model reaction to reveal the significant impact of EDL structure on CO adsorption. By altering the sequence of applying negative potential and elevating CO pressure, we discern two distinct EDL structures with varying cation density and CO coverage. Our findings demonstrate that the EDL comprising densely packed cations substantially hinders CO adsorption on the Cu as opposed to the EDL containing less compact cations. These two different EDL structures remained stable over the course of our experiments, despite their identical initial and final conditions, suggesting an insurmountable kinetic barrier present in between. Moreover, we show that the size and identity of cations play decisive roles in determining the properties of the EDL in CO electroreduction on Cu. This study presents a refined adaptation of the classical Gouy-Chapman-Stern model and highlights its catalytic importance, which bridges the mechanistic gap between the EDL structure and cathodic reactions.

20.
Nanoscale ; 16(8): 4095-4104, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38333905

RESUMO

In recent years, tumor catalytic therapy based on nanozymes has attracted widespread attention. However, its application is limited by the tumor hypoxic microenvironment (TME). In this study, we developed oxygen-supplying magnetic bead nanozymes that integrate hemoglobin and encapsulate the photosensitizer curcumin, demonstrating reactive oxygen species (ROS)-induced synergistic breast cancer therapy. Fe3O4 magnetic bead-mediated catalytic dynamic therapy (CDT) generates hydroxyl radicals (˙OH) through the Fenton reaction in the tumor microenvironment. The Hb-encapsulated Fe3O4 magnetic beads can be co-loaded with the photosensitizer/chemotherapeutic agent curcumin (cur), resulting in Fe3O4-Hb@cur. Under hypoxic conditions, oxygen molecules are released from Fe3O4-Hb@cur to overcome the TME hypoxia, resulting in comprehensive effects favoring anti-tumor responses. Upon near-infrared (NIR) irradiation, Fe3O4-Hb@cur activates the surrounding molecular oxygen to generate a certain amount of singlet oxygen (1O2), which is utilized for photodynamic therapy (PDT) in cancer treatment. Meanwhile, we validated that the O2 carried by Hb significantly enhances the intracellular ROS level, intensifying the catalytic therapy mediated by Fe3O4 magnetic beads and inflicting lethal damage to cancer cells, effectively inhibiting tumor growth. Therefore, significant in vivo synergistic therapeutic effects can be achieved through catalytic-photodynamic combination therapy.


Assuntos
Neoplasias da Mama , Curcumina , Neoplasias , Fotoquimioterapia , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Oxigênio , Espécies Reativas de Oxigênio/farmacologia , Curcumina/farmacologia , Curcumina/uso terapêutico , Linhagem Celular Tumoral , Fotoquimioterapia/métodos , Neoplasias/tratamento farmacológico , Hipóxia , Fenômenos Magnéticos , Microambiente Tumoral , Peróxido de Hidrogênio/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA