Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Fish Shellfish Immunol ; 154: 109901, 2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39276815

RESUMO

SOCS family genes are a class of repressors in various signaling pathways of mammals involved in regulating immunity, growth, and development, but the information remains limited in teleost. The full-length cDNA sequence of the Japanese eel SOCS6 gene, named AjSOCS6, was first cloned and showed to encode 529 amino acids with a conserved SH2 structural domain and a typical structure of a C-terminal SOCS box. AjSOCS6 is evolutionarily close to that of rainbow trout and zebrafish. AjSOCS6 gene expression was observed across all tissues in Japanese eel, with the highest levels found in the intestine. In vivo studies showed that AjSOCS6 was significantly upregulated in the liver following exposure to LPS, poly I:C, and Aeromonas hydrophila infection. In vitro, stimulation with poly I:C, CpG, and A. hydrophila infection increased AjSOCS6 expression in Japanese eel liver cells. Subcellular localization revealed that AjSOCS6 was dispersed in the cytoplasm. Overexpressing AjSOCS6 significantly suppressed the expression of immune-related genes, such as c-Rel and p65 in the NF-κB pathway, IFN1, IFN2, and IFN4 in the type I IFN signaling pathway, and the downstream inflammatory factor IL-6 in Japanese eel liver cells. Conversely, knocking down AjSOCS6 in vitro in liver cells and in vivo in the liver, spleen, and kidney significantly upregulated these gene expressions. Co-transfection of AjSOCS6 with AjMyD88 into HEK293 cells significantly reduced NF-κB luciferase activities compared to AjMyD88 single-transfection groups, in a natural state and under LPS stimulation. These findings suggest that AjSOCS6 negatively regulates MyD88-dependent NF-κB and type I IFN signaling pathways, underscoring its role in the immune defense of fish against viral and bacterial infections.

2.
Turk J Med Sci ; 53(3): 630-639, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37476905

RESUMO

BACKGROUND: Exosomes derived from oral squamous cell carcinoma (OSCC) could modulate OSCC development. This study aimed to explore effects of exosome-mediated lncRNA PART1 on OSCC cells. METHODS: This study was performed in Tianjin Medical University Cancer Institute from February 2021 to March 2022. Bioinformatic analysis was performed on the public database GEPIA (http://gepia.cancer-pku.cn/). Exosomes isolated from cell lines squamous cell carcinoma 9 (SCC9) and Centre Antoine Lacassagne-27 (CAL27) were identified by transmission electron microscope and western blot. Exosome-mediated lncRNA PART1, microRNA-17-5p(miR-17-5p) and suppressor of cytokine signaling 6(SOCS6) RNA expressions were assessed by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Cell counting kit8(CCK-8), caspase-3 activity, and flow cytometry were applied to evaluate OSCC cell viabilities and apoptosis. Meanwhile, OSCC cell migratory ability and invasiveness were evaluated using transwell assay. Bindings between miR-17-5p and lncRNA PART1 or SOCS6 were validated using the luciferase reporter test. Western blot was used for detecting the protein levels of SOCS6, phosphorylated signal transducer and activator of transcription 3 (STAT3) and STAT3. RESULTS: : According to GEPIA, lncRNA PART1 was downregulated in OSCC tissue samples and cells, and it had a positive correlation with the good prognosis of Head and neck squamous cell cancer (HNSCC) patients. After the exosomes from OSCC cells were isolated and verified, PART1 was then confirmed to be secreted by exosomes. Further, overexpression of exosome-mediated lncRNA PART1 inhibited OSCC cell viabilities, migration, and invasiveness but facilitated OSCC cell apoptosis. PART1 upregulated SOCS6 through sponging miR-17-5p. Moreover, exosome-mediated lncRNA PART1 suppressed the phosphorylation of STAT3. DISCUSSION: Exosome-mediated lncRNA PART1 could mediate the OSCC progression via miR-17-5p/SOCS6 axis in vitro, suggesting that lncRNA PART1 might be a target for treating OSCC.


Assuntos
Carcinoma de Células Escamosas , Exossomos , Neoplasias de Cabeça e Pescoço , MicroRNAs , Neoplasias Bucais , RNA Longo não Codificante , Humanos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , RNA Longo não Codificante/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Exossomos/genética , Exossomos/metabolismo , Exossomos/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Bucais/genética , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
3.
Biochem Biophys Res Commun ; 550: 92-98, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33689885

RESUMO

Esophageal cancer is one of the most common cancer of the digestive system and radiotherapy is widely applied in advanced esophageal cancer treatment, however radioresistance (RR) is one of the major reasons for radiotherapy failure. There is limited knowledge on the mechanisms that cause RR, here we identify suppressors of cytokine signaling 6 (SOCS6) is a negative regulator of radioresistance in ESCC cells. SOCS6 deficiency in ESCC cells conferred radioresistance in vitro and in vivo by increasing radiation-induced G2/M arrest, DNA damage repair and inhibiting radiation-induced apoptosis. Moreover, the transcriptome sequencing analysis demonstrates that the transcription of SOCS6 was partially p53-dependent. Importantly we found that highly correlated SOCS6 and P53 express lower in RR esophageal cancer tissues compare with radiosensitive ones. Collectedly our study uncovers that SOCS6, as a downstream effector of p53, is a key regulator involved in the radioresistance of ESCC.


Assuntos
Neoplasias Esofágicas/radioterapia , Carcinoma de Células Escamosas do Esôfago/radioterapia , Tolerância a Radiação/genética , Tolerância a Radiação/efeitos da radiação , Proteínas Supressoras da Sinalização de Citocina/deficiência , Proteínas Supressoras da Sinalização de Citocina/genética , Apoptose/efeitos da radiação , Linhagem Celular , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos da radiação , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos da radiação , Regiões Promotoras Genéticas/genética , Interferência de RNA , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Proteína Supressora de Tumor p53/metabolismo
4.
Cancer Cell Int ; 21(1): 165, 2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33712005

RESUMO

BACKGROUND: Radiotherapy is a major treatment for esophageal squamous cell carcinoma (ESCC). However, HPV infection related radioresistance caused poor prognosis of ESCC. The function of SOCS6, which has been shown to be a tumor suppressor in several cancers, has not been fully investigated up till now. In this manuscript, we aim to further investigate the role of SOCS6 in regulating ESCC radioresistance. METHODS: Fifty-seven ESCC patients were enrolled for survival analysis. SOCS6 was stably overexpressed in HPV+ ESCC and ESCC cells, and cells were treated with radiation and then subjected to colony formation assays. Expression of DNA damage repair regulating proteins were examined by Western blotting. Cell growth, cell migration and cisplatin sensitivity were then analyzed. Sphere formation assays and flow cytometry were used to investigate changes in cancer stem cell (CSC) properties. Immunofluorescent staining and confocal microscopy were used to locate SOCS6 and c-Kit. Ubiquitylation level of c-Kit were analyzed after immunoprecipitation. Then, coimmunoprecipitation (CoIP) of SOCS6 and c-Kit were performed. In vivo, xenograft animal models were treated with radiation to examine the radiosensitivity. RESULTS: SOCS6 is correlated with better prognosis in ESCC patients. Radioresistance is impaired by SOCS6 upregulation, which inhibited cell growth, migration and increased sensitivity to cisplatin. SOCS6 significantly decreased the population of CSCs expressing the surface biomarker CD271 or CD24low/CD44high and their ability of sphere formation. SOCS6 and c-Kit were collocated in the cytoplasm. Blotting of ubiquitin and CoIP experiments indicated that the mechanism was related to ubiquitylation and degradation of the receptor c-Kit. Xenograft tumor mouse model showed that SOCS6 inhibited tumor growth and promoted radiosensitivity in vivo. CONCLUSIONS: Our findings suggest that SOCS6 can promote the radiosensitivity of HPV+ ESCC and ESCC cells and reduce their stemness via ubiquitylation and degradation of c-Kit. Thus, SOCS6 is a potential target for overcoming radioresistance of ESCC.

5.
Exp Cell Res ; 390(1): 111955, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32165166

RESUMO

Tumor-initiating cells (T-ICs) are involved in the tumorigenesis, progression, drug resistance and recurrence of hepatocellular carcinoma (HCC). However, the underlying mechanism for the propagation of liver T-ICs remains unclear. Herein, we find that miR-454 is upregulated in liver T-ICs and has an important function in liver T-ICs. Functional studies have revealed that knockdown of miR-454 inhibits liver T-IC self-renewal and tumorigenesis. Conversely, forced miR-454 expression promotes liver T-IC self-renewal and tumorigenesis. Mechanistically, we found that miR-454 downregulates SOCS6 expression in liver T-ICs. The correlation between miR-454 and SOCS6 is validated in human HCC tissues. Furthermore, HCC cells that overexpress miR-454 are resistant to sorafenib treatment. Analysis of patient-derived xenografts (PDXs) further demonstrates that miR-454 may predict sorafenib benefits in HCC patients. In conclusion, our findings reveal the crucial role of miR-454 in liver T-IC expansion and sorafenib response.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Animais , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Células Hep G2 , Humanos , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , MicroRNAs/genética , Células-Tronco Neoplásicas/fisiologia , Sorafenibe/uso terapêutico , Proteínas Supressoras da Sinalização de Citocina/metabolismo
6.
Int J Mol Sci ; 21(14)2020 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-32668737

RESUMO

The BC-box motif in suppressor of cytokine signaling 6 (SOCS6) promotes the neuronal differentiation of somatic stem cells, including epidermal stem cells. SOCS6 protein belongs to the group of SOCS proteins and inhibits cytokine signaling. Here we showed that epidermal stem cells were induced to differentiate into GABAnergic neurons by the intracellular delivery of a peptide composed of the amino-acid sequences encoded by the BC-box motif in SOCS6 protein. The BC-box motif (SLQYLCRFVI) in SOCS6 corresponded to the binding site of elongin BC. GABAnergic differentiation mediated by the BC-box motif in SOCS6 protein was caused by ubiquitination of JAK2 and inhibition of the JAK2-STAT3 pathway. Furthermore, GABAnergic neuron-like cells generated from epidermal stem cells were transplanted into the brain of a rodent ischemic model. Then, we demonstrated that these transplanted cells were GAD positive and that the cognitive function of the ischemic model rodents with the transplanted cells was improved. This study could contribute to not only elucidating the mechanism of GABAnergic neuronal differentiation but also to neuronal regenerative medicine utilizing GABAnergic neurons.


Assuntos
Células Epidérmicas/efeitos dos fármacos , Neurônios GABAérgicos/citologia , Neurogênese/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/farmacologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células Cultivadas , Transtornos Cognitivos/etiologia , Células Epidérmicas/citologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/cirurgia , Janus Quinase 2/metabolismo , Microscopia de Fluorescência , Teste do Labirinto Aquático de Morris , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Células-Tronco Pluripotentes/transplante , Processamento de Proteína Pós-Traducional , Ratos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/administração & dosagem , Proteínas Supressoras da Sinalização de Citocina/química , Ubiquitinação
7.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1312-1325, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29932990

RESUMO

Hypothalamic leptin receptor (LR) signaling regulates body weight by balancing food intake and energy expenditure. It is well established that the human LR undergoes ectodomain shedding, but little is known about the fate of the remaining cytosolic domain. This study demonstrates that regulated intramembrane proteolysis (RIP) releases the LR intracellular domain (LR ICD), which translocates to the mitochondria where it binds to SOCS6. This LR ICD-SOCS6 interaction stabilizes both proteins on the mitochondrial outer membrane and requires a functional BC box in SOCS6 for mitochondrial association and a central motif in the LR ICD for SOCS6 binding. The LR ICD prevents CCCP-induced mitochondrial depolarization and mitophagy as shown by lowered Parkin translocation and p62 accumulation. Strict regulation of mitochondrial dynamics in the hypothalamus is known to be essential for body weight homeostasis. This is the first study showing that the LR can directly modulate mitochondrial biology.


Assuntos
Mitocôndrias/fisiologia , Receptores para Leptina/química , Receptores para Leptina/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Linhagem Celular Tumoral , Polaridade Celular/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Hipotálamo/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitofagia , Domínios Proteicos , Proteólise
8.
IUBMB Life ; 71(7): 882-890, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30809930

RESUMO

Oral squamous cell carcinoma (OSCC) is a lethal malignancy and its prognosis remains dismal. Thus, a deeper understanding of the mechanisms is needed to provide a new insight for new therapies. It has been reported that long noncoding RNA (lncRNA) maternally expressed gene 3 (MEG3) was downregulated in OSCC tissues, however, its functional mechanism remains uncertain. Here, we found that the overexpression of MEG3 suppressed migration and promoted apoptosis in OSCC cell lines, while inhibition of MEG3 exhibited opposite effect. We also found that MEG3 could effectively sponge miR-548d-3p and decrease its expression level. Moreover, miR-548d-3p repressed the expression of SOCS5 and SOCS6 through binding their 3'UTR, thereby modulating the JAK-STAT signaling pathway and functioning as an oncogene in OSCC cells. Importantly, overexpression of MEG3 enhanced the expression of SOCS5 and SOCS6 to regulate JAK-STAT pathway, whereas miR-548d-3p overexpression decreased the effects of MEG3 on levels of SOCS5/SOCS6. Furthermore, upregulated expression of miR-548d-3p could abrogate the effect of MEG3 overexpression on migration and apoptosis in OSCC cell lines. In addition, the overexpression of MEG3 inhibited tumor migration and facilitated apoptosis in vivo. Together, our results revealed that MEG3 could modulate JAK-STAT pathway via miR-548d-3p/SOCS5/SOCS6 to suppresses migration and promote apoptosis in OSCC. Our research indexed a new functional mechanism of MEG3 in OSCC, and this mechanism may be a potential prognostic factor and therapeutic target. © 2019 IUBMB Life, 2019.


Assuntos
Apoptose , Carcinoma de Células Escamosas/patologia , Movimento Celular , Janus Quinase 1/metabolismo , MicroRNAs/genética , Neoplasias Bucais/patologia , RNA Longo não Codificante/genética , Fator de Transcrição STAT3/metabolismo , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Janus Quinase 1/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Fator de Transcrição STAT3/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cell Physiol Biochem ; 49(6): 2382-2395, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30261490

RESUMO

BACKGROUND/AIMS: Glioblastoma is the most common and aggressive brain tumor and carries a poor prognosis. Previously, we found that neurotensin receptor 1 (NTSR1) contributes to glioma progression, but the underlying mechanisms of NTSR1 in glioblastoma invasion remain to be clarified. The aim of this study was to investigate the molecular mechanisms of NTSR1 in glioblastoma invasion. METHODS: Cell migration and invasion were evaluated using wound-healing and transwell assays. Cell proliferation was detected using CCK-8. The expression of NTSR1, Jun, and suppressor of cytokine signaling 6 (SOCS6) was detected using western blotting. The expression of miR-494 was detected by Quantitative real-time PCR. Chromatin immunoprecipitation assay was performed to examine the interaction between Jun and miR-494 promoter. Dual-luciferase reporter assay and western blotting were performed to identify the direct regulation of SOCS6 by miR-494. An orthotopic xenograft mouse model was conducted to assess tumor growth and invasion. RESULTS: NTSR1 knockdown attenuated the invasion of glioblastoma cells. Jun was positively regulated by NTSR1, which promoted miR-494 expression through binding to miR-494 promoter. SOCS6 was confirmed as a direct target of miR-494, thus, NTSR1-induced miR-494 upregulation resulted in SOCS6 downregulation. Both miR-494 and SOCS6 were involved in the NTSR1-induced invasion of glioblastoma cells. In vivo, tumor invasion and growth were inhibited by NTSR1 knockdown, but were restored with miR-494 overexpression. CONCLUSION: NTSR1 knockdown inhibited glioblastoma invasion via the Jun/miR-494/SOCS6 axis.


Assuntos
MAP Quinase Quinase 4/metabolismo , MicroRNAs/metabolismo , Receptores de Neurotensina/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Regiões 3' não Traduzidas , Animais , Antagomirs/metabolismo , Encéfalo/diagnóstico por imagem , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Glioblastoma/diagnóstico por imagem , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , MAP Quinase Quinase 4/genética , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico , Receptores de Neurotensina/antagonistas & inibidores , Receptores de Neurotensina/genética , Proteínas Supressoras da Sinalização de Citocina/química , Proteínas Supressoras da Sinalização de Citocina/genética
10.
Biochem Biophys Res Commun ; 495(1): 1363-1369, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28986253

RESUMO

microRNAs (miRNAs) play critical roles in cancer development and progression. This study investigated the effects of miR-19 in human osteosarcoma (OS) development. Here, we showed that miR-19 was frequently upregulated in OS tissues and cell lines. Moreover the expression of miR-19 was associated with TNM stage, metastasis, size and poor overall survival. Mechanistically, miR-19 dramatically suppressed OS growth in vitro and in vivo. Bioinformatics analyses predicted that SOCS6 is a potential target gene of miR-19 in OS, which was confirmed by luciferase-reporter assay. We also found that SOCS6 expression was downregulated and negatively correlated with miR-19 expression in OS tissues clinically. Moreover, ectopic SOCS6 could reverse miR-19 induced OS growth. Finally, JAK2/STAT3 signaling pathway involves miR-19/SOCS6-mediated OS progression. Together, our data provide important evidence for miR-19 mediated SOCS6 in OS growth and revealed miR-19/SOCS6/JAK2/STAT3 pathway as a potential therapeutic strategy for OS patients.


Assuntos
Proliferação de Células , MicroRNAs/metabolismo , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia , Regulação para Cima
11.
Mol Carcinog ; 56(12): 2610-2619, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28731203

RESUMO

PTEN pseudogene (PTENP1) has a tumor suppressive role in multiple cancers. However, its involvement in esophageal squamous cell carcinoma (ESCC) remains largely unknown. In this study, we set out to identify the role of PTENP1 in the development of ESCC. Gene Expression Omnibus database was employed to investigate the expression of PTENP1 in ESCC. sRNA target Database (StarBase v2.0) was used to query the downstream of PTENP1. Next, both in vitro and in vivo experiments were employed to explore the function. Cell proliferation was evaluated by CCK-8, soft agar, and colony formation assays. Expression of relative genes was assessed by quantitative real-time PCR (qRT-PCR) and Western blotting. 3'UTR luciferase assay was used to confirm the miRNA binding. The clinical significance of PTENP1 was further validated by immunohistochemistry (IHC) and correlation with clinicopathological indicators in additional samples (n = 93). We found expression of PTENP1 in ESCC was lower than that in the corresponding adjacent normal tissues (n = 17). Overexpression of PTENP1 in Eca109 and TE-1 cells resulted in inhibited proliferation and altered expression of SOCS6-p-STAT3-HIF-1α pathway both in vitro and in vivo. Subsequent IHC reported a similar trend in human ESCC samples. 3'UTR luciferase assay demonstrated that PTENP1 3'UTR decoyed miR-17-5p from binding to SOCS6. Moreover, PTENP1 expression was correlated with clinicopathological indicators to varying degrees, including histological grade, TNM stage, infiltration depth, lymph node metastasis, and overall survival. Taken together, these results suggested an anti-oncogenic role of PTENP1. Meanwhile, PTENP1 may also serve as a candidate of prognostic indicator for ESCC patients.


Assuntos
Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Regulação Neoplásica da Expressão Gênica , PTEN Fosfo-Hidrolase/genética , Pseudogenes , Proteínas Supressoras da Sinalização de Citocina/genética , Regiões 3' não Traduzidas/genética , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Feminino , Células HEK293 , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , PTEN Fosfo-Hidrolase/metabolismo , Interferência de RNA , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Transplante Heterólogo
12.
Int Immunopharmacol ; 130: 111638, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38373387

RESUMO

L-arginine, as an essential substance of the immune system, plays a vital role in innate immunity. MiR155, a multi-functional microRNA, has gained importance as a regulator of homeostasis in immune cells. However, the immunoregulatory mechanism between L-arginine and miR155 in bacterial infections is unknown. Here, we investigated the potential role of miR155 in inflammation and the molecular regulatory mechanisms of L-arginine in Streptococcus uberis (S. uberis) infections. And we observed that miR155 was up-regulated after infection, accompanying the depletion of L-arginine, leading to metabolic disorders of amino acids and severe tissue damage. Mechanically, the upregulated miR155 mediated by the p65 protein played a pro-inflammatory role by suppressing the suppressor of cytokine signaling 6 (SOCS6)-mediated p65 ubiquitination and degradation. This culminated in a violently inflammatory response and tissue damage. Interestingly, a significant anti-inflammatory effect was revealed in L-arginine supplementation by reducing miR155 production via inhibiting p65. This work firstly uncovers the pro-inflammatory role of miR155 and an anti-inflammatory mechanism of L-arginine in S.uberis infection with a mouse mastitis model. Collectively, we provide new insights and strategies for the prevention and control of this important pathogen, which is of great significance for ensuring human food health and safety.


Assuntos
Arginina , Mastite , MicroRNAs , Infecções Estreptocócicas , Animais , Feminino , Humanos , Camundongos , Arginina/metabolismo , Inflamação/metabolismo , MicroRNAs/genética , Infecções Estreptocócicas/metabolismo , Streptococcus/fisiologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Mastite/imunologia , Mastite/metabolismo
13.
Adv Clin Exp Med ; 32(8): 873-880, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37589228

RESUMO

BACKGROUND: Diabetic retinopathy (DR) is one of the most common microvascular complications of diabetes mellitus (DM). A recent in vitro study found that the suppressor of cytokine signaling 6 (SOCS6) plays a protective role in DR and DM. However, to date, no clinical studies have focused on the role of SOSC6 in DR development. OBJECTIVES: The present study aimed to investigate the expression and clinical significance of serum SOCS6 in DR. MATERIAL AND METHODS: A total of 159 DR patients were enrolled in the study. Additionally, 156 type 2 DM (T2DM) patients without DR were recruited as controls. Serum levels of SOCS6, C-reactive protein (CRP), interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), vascular endothelial growth factor (VEGF), and angiopoietin-2 (ANG-2) were measured using enzyme-linked immunosorbent assay (ELISA). Demographic and clinical data were collected. RESULTS: Age, the course of DM, systolic blood pressure (SBP), diastolic blood pressure (DBP), and the levels of low-density lipoprotein cholesterol (LDL-C) and total cholesterol (TC) were significantly higher in proliferative DR (PDR) patients. Serum SOCS6 levels in PDR patients were remarkably lower than in non-PDR patients or non-DR T2DM patients. The Pearson's analysis showed that SOCS6 was negatively correlated with CRP, IL-6, TNF-α, IL-1ß, VEGF, and ANG-2. The serum levels of CRP, IL-6, TNF-α, IL-1ß, VEGF, and ANG-2 in the SOCS6 low expression group were significantly increased compared to patients with high SOCS6 levels. Receiver operating characteristic (ROC) curves showed that SOCS6 could be a potential diagnostic biomarker for DR. For logistic regression, 3 models were used. It was found that SOCS6, the course of DM, SBP and DBP in model 1, IL-1ß and TNF-α in model 2, and VEGF and ANG-2 in model 3 were risk factors for DR. CONCLUSIONS: The SOCS6 is decreased in DR patients and is related to severity and clinical outcomes, including inflammatory and angiogenic factors.


Assuntos
Diabetes Mellitus Tipo 2 , Retinopatia Diabética , Humanos , Retinopatia Diabética/etiologia , Fator A de Crescimento do Endotélio Vascular , Interleucina-6 , Fator de Necrose Tumoral alfa , Diabetes Mellitus Tipo 2/complicações , Proteína C-Reativa , LDL-Colesterol , Proteínas Supressoras da Sinalização de Citocina
14.
Innate Immun ; 28(5): 174-184, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35678490

RESUMO

Periodontitis is a prevalent chronic inflammatory disease. Circular RNAs (circRNAs) have been revealed to play roles in the inflammatory response. Hence, this work aimed to explore the role and mechanism of circ_0062491 in periodontitis progression. Human periodontal ligament cells (PDLCs) were isolated from the periodontal ligament (PDL) of the healthy teeth with orthodontic requirement after tooth extraction. In vitro experiments were conducted by cell counting Kit-8 (CCK-8) assay, flow cytometry, Western blot, and ELISA to determine cell viability, apoptosis, and inflammatory response. The binding between miR-498 and circ_0062491 or SOCS6 was confirmed using dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. Circ_0062491 expression was decreased in periodontitis and LPS-induced PDLCs. Restoration of circ_0062491 attenuated LPS-induced apoptosis and inflammation in PDLCs in vitro. Mechanistically, circ_0062491 functioned as a sponge for miR-498, and miR-498 directly targeted SOCS6. Rescue experiments showed that miR-498 up-regulation reversed the protective action of circ_0062491 on PDLCs under LPS treatment. Moreover, silencing of miR-498 protected PDLCs from LPS-induced apoptosis and inflammation, which were abolished by SOCS6 knockdown. Circ_0062491 protected PDLCs from LPS-induced apoptosis and inflammation, suggesting a new target for the amelioration of periodontitis patients.


Assuntos
MicroRNAs , Periodontite , Apoptose/genética , Humanos , Inflamação/genética , Lipopolissacarídeos , MicroRNAs/genética , MicroRNAs/metabolismo , Periodontite/genética , Periodontite/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
15.
Cytokine Growth Factor Rev ; 64: 21-32, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35210167

RESUMO

The suppressor of cytokine signaling 6 (SOCS6), an emerged important member of SOCS family, has attracted enhancing attention regarding its pivotal role in the development and progression of cancers. As part of negative feedback regulation, SOCS6 has been implicated in attenuating cytokine signal transduction via inhibiting the signaling cascade of activated cytokine receptors and multiple receptor tyrosine kinase signaling. Decreased SOCS6 expression is involved in diverse tumorigenic processes, such as abnormal cell proliferation, evasion of apoptosis, cancer migration, and cancer stem cell maintenance. Herein, this review summarized the mechanisms of SOCS6 regulation underlying multiple pathways. In particular, we focus on the pathological processes of cancer targeted by SOCS6 and discuss its inhibitory role during tumor progression. Also, we focused on the clinical relevance of SOCS6 in cancer biomarker and prognosis, as well as its significance in chemoresistance and radioresistance. In all, this review pave a way to assist in experimental designs and emphasize the potential clinical value of SOCS6 for cancer.


Assuntos
Neoplasias , Proteínas Supressoras da Sinalização de Citocina , Apoptose , Citocinas/metabolismo , Humanos , Neoplasias/terapia , Transdução de Sinais/fisiologia , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
16.
Inflammation ; 45(5): 1936-1949, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35461393

RESUMO

The objective of this study is to investigate lncRNA FLG-AS1-mediated miR-380-3p/SOCS6 axis in inflammation, oxidative stress, and apoptosis of retinal epithelial cells in diabetic retinopathy (DR). Fasting blood was collected from 60 DR patients and 60 healthy controls. The Pearson correlation was used to analyze the correlation between the expression levels of FLG-AS1 and miR-380-3p in DR patients. qRT-PCR and/or Western blotting were used to detect the expression of FLG-AS1, miR-380-3p, and SOCS6. After gain of function of FLG-AS1 or SOCS6 or loss of function of miR-380-3p, high glucose (HG)-treated human retinal pigment epithelial ARPE-19 cells were subjected to TUNEL assessment of apoptosis. ELISA was performed to detect the expression levels of IL-1ß, IL-6, and TNF-α in cell culture supernatant. DCFH-DA was used to detect the level of ROS in the cells. MDA and SOD assay kits were used to measure the activity of MDA and SOD in the cells. Dual-luciferase reporter assay was performed to verify the binding between miR-380-3p and FLG-AS1 or between miR-380-3p and SOCS6. Streptozotocin injections were used to induce diabetes in rats which were injected with FLG-AS1 overexpression lentiviral vectors in the eye. Twenty weeks later, retinal tissue was isolated and stained with hematoxylin-eosin or TUNEL. Compared to that in healthy controls, FLG-AS1 expression decreased 2.5-fold and miR-380-3p expression increased 2.6-fold in the serum of DR patients. The expression levels of FLG-AS1 and miR-380-3p were negatively correlated in DR patients (r = -0.3772, P = 0.003). Overexpression of FLG-AS1 reduced inflammation, oxidative stress, and apoptosis of HG-treated ARPE-19 cells and alleviated retinal injury in diabetic rats. FLG-AS1 promoted the expression of SOCS6 by targeting miR-380-3p. Inhibition of miR-380-3p or overexpression of SOCS6 reduced inflammation, oxidative stress, and apoptosis of HG-treated ARPE-19 cells. FLG-AS1 mitigates DR by regulating retinal epithelial cell inflammation, oxidative stress, and apoptosis via the miR-380-3p/SOCS6 axis.


Assuntos
Diabetes Mellitus Experimental , Retinopatia Diabética , MicroRNAs , RNA Longo não Codificante , Animais , Apoptose , Proliferação de Células , Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/genética , Retinopatia Diabética/metabolismo , Amarelo de Eosina-(YS)/metabolismo , Células Epiteliais/metabolismo , Glucose/metabolismo , Hematoxilina/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-6/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Estresse Oxidativo , RNA Longo não Codificante/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Pigmentos da Retina/metabolismo , Estreptozocina/metabolismo , Superóxido Dismutase/metabolismo , Proteínas Supressoras da Sinalização de Citocina , Fator de Necrose Tumoral alfa/metabolismo
17.
Front Med (Lausanne) ; 9: 988962, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36341243

RESUMO

Numerous microRNAs (miRNAs) have been found to have an aberrant expression in the peripheral blood or psoriasis patients' lesions. Psoriasis was shown to have the abnormal expression of microRNA-203 (miR-203). It is a skin-specific signal that governs cellular proliferation in a protein kinase C-dependent manner and is mostly generated by keratinocytes. This work evaluated the expression levels of the circulating miR-203 target genes SOCS3, SOCS6, TP63, TNF-, IL8, and IL24 in psoriasis patients. Using a relative quantitation PCR technique, we determined the expression levels of miR-203 and its target genes (SOCS3, SOCS6, TP63, TNF-, IL8, and IL24) in the plasma of 120 psoriatic patients and matched healthy controls. The disease characteristics of the patients were then correlated with the expression results. We also conducted numerous enrichment analyses for the diseases, functions, and pathways connected to the under-researched biomarkers. Compared to healthy controls, psoriatic patients had significantly increased levels of miR-203 expression; 7.1 (4.4-9.9). In contrast, psoriatic patients had significantly lower expression of all the examined genes compared to healthy controls. Regarding all the study biomarkers, the receiver operating characteristic (ROC) curve analysis demonstrated significant sensitivity and specificity for differentiating between psoriatic patients and healthy controls. According to the results of the disease matching score generated by miR-203 and its target genes, psoriasis was ranked first with a score of 4.45. The third-place finisher with a value of 3.98, it also demonstrated that miR-203 and its target genes are connected to various skin disorders. Our results show that miR-203 contributes to psoriasis pathogenesis not only locally in skin lesions but also in circulation, indicating that it may contribute to the systemic symptoms of the illness. MiR-203 overexpression in psoriasis suggests that miR-203 may be involved in an anti-inflammatory response because it targets both SOCS gene family members and pro-inflammatory cytokines.

18.
J Cancer Res Clin Oncol ; 148(12): 3485-3499, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35907010

RESUMO

OBJECTIVE: Lung cancer (LC) remains a threatening health issue worldwide. Methyltransferase-like protein 3 (METTL3) is imperative in carcinogenesis via m6A modification of microRNAs (miRNAs). This study estimated the effect of METTL3 in LC by regulating m6A methylation-mediated pri-miR-663 processing. METHODS: miR-663 expression in 4 LC cell lines and normal HBE cells was determined using RT-qPCR. A549 and PC9 LC cells selected for in vitro studies were transfected with miR-663 mimics or inhibitor. Cell viability, migration, invasion, proliferation, and apoptosis were detected by CCK-8, Transwell, EdU, and flow cytometry assays. The downstream target genes and binding sites of miR-663 were predicted via Starbase database and validated by dual-luciferase assay. LC cells were delivered with oe-METTL3/sh-METTL3. Crosslinking between METTL3 and DGCR8 was verified by co-immunoprecipitation. Levels of m6A, miR-663, and pri-miR-663 were measured by m6A dot blot assay and RT-qPCR. m6A modification of pri-miR-663 was verified by Me-RIP assay. Finally, the effects of METTL3 in vivo were ascertained by tumor xenograft in nude mice. RESULTS: miR-663 was upregulated in LC cells, and miR-663 overexpression promoted cell proliferation, migration, invasion, and inhibited apoptosis, but miR-663 knockdown exerted the opposite effects. miR-663 repressed SOCS6 expression. SOCS6 overexpression annulled the promotion of miR-663 on LC cell growth. METTL3 bound to DGCR8, and METTL3 silencing elevated the levels of pri-miR-663 and m6A methylation-modified pri-miR-663, and suppressed miR-663 maturation and miR-663 expression. METTL3 facilitated tumor growth in mice through the miR-663/SOCS6 axis. CONCLUSION: METTL3 promotes LC progression by accelerating m6A methylation-mediated pri-miR-663 processing and repressing SOCS6.


Assuntos
Neoplasias Pulmonares , MicroRNAs , Humanos , Animais , Camundongos , Metiltransferases/genética , Metiltransferases/metabolismo , Metilação , MicroRNAs/genética , MicroRNAs/metabolismo , Camundongos Nus , Sincalida/metabolismo , Proteínas de Ligação a RNA/metabolismo , Neoplasias Pulmonares/genética , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
19.
Genes Dis ; 9(3): 697-704, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35782970

RESUMO

This study investigated the role of N6-methyladenosine RNA methylation in liver regeneration following partial hepatectomy in mice. We created a liver-specific knockout mouse model by the deletion of Mettl3, a key component of the N6-methyladenosine methyltransferase complex, using the albumin-Cre system. Mettl3 liver-specific knockout mice and their wild-type littermates were subjected to 2/3 partial hepatectomy. Transcriptomic changes in liver tissue at 48 h after partial hepatectomy were detected by RNA-seq. Immunohistochemistry and immunofluorescence were used to determine protein expression levels of Ki67, hepatocyte nuclear factor 4 alpha, and cytokeratin 19. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling was also performed. Liver weight/body weight ratios after partial hepatectomy were significantly lower in Mettl3 liver-specific knockout mice than in wild-type mice at 48 h after 2/3 partial hepatectomy (3.1% ± 0.11% vs. 2.7% ± 0.03%). Compared with wild-type littermates, Mettl3 liver-specific knockout mice showed reduced bromodeoxyuridine staining and reduced Ki-67 expression at 48 h after 2/3 partial hepatectomy. RNA-seq analysis showed that Mettl3 liver-specific knockout delayed the cell cycle progression in murine liver by downregulating the expression levels of genes encoding cyclins D1, A2, B1, and B2. Loss of Mettl3-mediated N6-methyladenosine function led to attenuated liver regeneration by altering the mRNA decay of suppressor of cytokine signaling 6, thereby inhibiting the phosphorylation of signal transducer and activator of transcription 3 during early liver regeneration. These results demonstrated the importance of N6-methyladenosine mRNA modification in liver regeneration and suggest that Mettl3 targeting might facilitate liver regeneration.

20.
Ann Transl Med ; 9(15): 1255, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34532392

RESUMO

BACKGROUND: Non-small cell lung cancer (NSCLC) is one of the most lethal malignancies worldwide. Deepening understanding of the pathogenesis of NSCLC is quite important for its treatment. Circular (circ) RNA_0015278 has been found to be downregulated in NSCLC, but its role in NSCLC and the underlying regulatory mechanism is unknown. METHODS: Circ_0015278, microRNA (miR)-1278 and SOCS6 were analyzed with real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) or western blot. Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2'-deoxyuridine (EdU) staining were used to evaluate cell proliferation. The colony forming capacity and invasion of NSCLC cells were assessed with colony formation and transwell assays, respectively. The interaction among circ_0015278, miR-1278, and SOCS6 was evaluated using luciferase, receptor interacting protein (RIP), and RNA-pull down assays. Cell apoptosis was analyzed using flow cytometry. A subcutaneous NSCLC xenograft mouse model was established for evaluating circ_0015278-mediated effects on the growth of NSCLC in vivo. RESULTS: Circ_0015278 was downregulated in NSCLC tissues and cells, and its reduced expression indicated poor prognosis. Overexpression of circ_0015278 restrained the proliferation, colony formation, invasion, and epithelial-mesenchymal transition (EMT) of NSCLC cells and induced NSCLC cell apoptosis. Moreover, overexpression of circ_0015278 inhibited the growth of NSCLC in vivo. Mechanically, circ_0015278 acted as an miR-1278 sponge to reduce its quantity, and miR-1278 targeted SOCS6 to inhibit its expression in NSCLC cells. Circ_0015278 promoted SOCS6 expression by sponging miR-1,278 in NSCLC cells. Overexpression of circ_0015278 attenuated the malignant phenotypes of NSCLC through sponging miR-1278 and consequently promoting SOCS6 expression. CONCLUSIONS: We demonstrated for the first time that circ_0015278 attenuated the progression of NSCLC via targeting the miR-1278/SOCS6 axis, which provides potential diagnostic biomarkers and therapeutic targets.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA