Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Arch Biochem Biophys ; 752: 109891, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38218360

RESUMO

Endometrial cancer (EC) is a common gynecological malignancy, and advanced-stage or recurrent EC is associated with a high mortality rate owing to the ineffectiveness of currently available treatments. FK506-binding protein 38 (FKBP38) is a member of the immunophilin family and inhibits melanoma and breast cancer cell metastasis. However, the functions of FKBP38 and its potential mechanism in EC remain unclear. Herein, we analyzed the expression levels of FKBP38 in EC cells and found that the FKBP38 expression was high in Ishikawa cells, and low in AN3CA cells, traditionally considered a low grade and a high grade cell line, respectively, in pathology classification. Moreover, FKBP38 inhibited cell proliferation, migration and invasion in EC cells, FKBP38 knockdown significantly promoted tumor growth of Ishikawa cells in a subcutaneous xenograft model and increased the number of lung metastases of Hec-1-A cells in a metastatic mouse model. Furthermore, FKBP38 suppressed several target proteins of epithelial-to-mesenchymal transition (EMT) and reduced the phosphorylation of ribosomal S6 protein (S6), eukaryotic initiation factor 4E-binding protein 1 (4EBP-1), indicating the potent inhibition of the mammalian target of rapamycin (mTOR) pathway. Meanwhile, the inhibition of mTOR neutralized the elevation of EC cell proliferation, migration and invasion after FKBP38 knockdown. In summary, FKBP38 would exert a tumor-suppressing role by modulating the mTOR pathway. Our results indicate that FKBP38 may be considered as a factor of EC metastasis and a new target for EC therapeutic intervention.


Assuntos
Neoplasias do Endométrio , Proteínas de Ligação a Tacrolimo , Animais , Feminino , Humanos , Camundongos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias do Endométrio/metabolismo , Mamíferos/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Ligação a Tacrolimo/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
J Pathol ; 257(2): 146-157, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35072951

RESUMO

Obesity is one of the major risk factors for cancer. Clinical studies have demonstrated that circulating levels of adiponectin are inversely correlated, not only with the extent of adiposity, but also with the incidence of several types of cancer, particularly endometrial cancer (EC). However, thus far, adiponectin remains a correlative factor, without definitive evidence to show a causal effect in EC and the potential mechanism(s) involved. To address this issue, we introduced an Apn-null mutation into Pten haploid-deficient (Pten+/- ) mice. The Pten heterozygous mutation alone led to the development of EC in less than 30% of female mice; however, when combined with the Apn-null mutation, the incidence of endometrial lesions rose to at least two-thirds. Although Apn deficiency did not further potentiate the Akt activation caused by the Pten mutation, it elevated the phosphorylation of mitogen-activated protein kinase (MAPK) p42/44, indicating activation of the MAPK signaling pathway. Treatment of Apn-/- ;Pten+/- mice with a MEK inhibitor blocked the development of EC. Finally, xenografts of a PTEN-proficient human EC cell line grew faster in Apn-deficient mice, whereas an adiponectin receptor agonist reduced xenograft growth of a PTEN-deficient human EC cell line. Thus, reduction of adiponectin activity promotes EC development, at least in the context of Pten mutation, by activating MAPK. © 2022 The Pathological Society of Great Britain and Ireland.


Assuntos
Neoplasias do Endométrio , Erros Inatos do Metabolismo , Adiponectina/deficiência , Adiponectina/genética , Adiponectina/farmacologia , Animais , Neoplasias do Endométrio/patologia , Feminino , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
3.
Invest New Drugs ; 40(5): 875-883, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35674866

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that frequently develops resistance to chemotherapy. A new approach to treating TNBC is required to improve patient survival. Phosphodiesterase-4 (PDE4) is an enzyme that is predominantly involved in the modulation of intracellular signaling mediated by cAMP. Although the efficacy of PDE4 inhibitors in several human inflammatory diseases is well documented, their clinical utility has been limited by side effects, including nausea and emesis. Recently, PDE4 has been used as a potential therapeutic target for different cancer types. In the present study, we investigated the anticancer effects of a novel PDE4 inhibitor ZL-n-91 on TNBC and the underlying mechanism. We showed that ZL-n-91 inhibited the proliferation of TNBC cells, induced cell apoptosis, and caused cell cycle arrest. Western blot analysis showed that ZL-n-91 increased Bax level and reduced Bcl-2 expression. Furthermore, downregulation of the cell cycle-related proteins, such as CDK2, CDK4, cyclin D1, PCNA, p-RB, and ZL-n-91, significantly inhibited the transcription of DNA repair genes and triggered an intracellular DNA damage response. Moreover, ZL-n-91 prevented the growth of the transplanted MDA-MB-231 tumor xenograft in nude mice and increased the γ-H2AX expression. These data demonstrate the anticancer effects of ZL-n-91 on TNBC cells and suggest its potential use in anticancer therapy.


Assuntos
Inibidores da Fosfodiesterase 4 , Neoplasias de Mama Triplo Negativas , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/farmacologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/uso terapêutico , Humanos , Camundongos , Camundongos Nus , Inibidores da Fosfodiesterase 4/farmacologia , Inibidores da Fosfodiesterase 4/uso terapêutico , Neoplasias de Mama Triplo Negativas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Diabetologia ; 64(5): 1169-1183, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33544171

RESUMO

AIMS/HYPOTHESIS: Besides serving as a traditional inflammatory marker, C-reactive protein (CRP) is closely associated with the development of obesity, diabetes and cardiovascular diseases as a metabolic and inflammatory marker. We hypothesise that CRP protein directly participates in the regulation of energy and glucose metabolism rather than just being a surrogate marker, and that genetic deficiency of CRP will lead to resistance to obesity and insulin resistance. METHODS: Crp gene deletion was achieved by transcription activator-like effector nuclease (TALEN) technology in rats. The Crp knockout animals were placed on either a standard chow diet or a high-fat diet. Phenotypic changes in body weight, glucose metabolism, insulin sensitivity, energy expenditure and inflammation condition were examined. The central impact of CRP deficiency on leptin and insulin hypothalamic signalling, as well as glucose homeostasis, were examined via intracerebral ventricular delivery of leptin and CRP plus glucose clamp studies in the wild-type and Crp knockout rats. RESULTS: CRP deficiency led to a significant reduction in weight gain and food intake, elevated energy expenditure and improved insulin sensitivity after exposure to high-fat diet. Glucose clamp studies revealed enhanced hepatic insulin signalling and actions. Deficiency of CRP enhanced and prolonged the weight-reducing effect of central injected leptin and promoted the central and peripheral roles of leptin. By contrast, reinstatement of CRP into the hypothalamus of the knockout rats attenuated the effects of central leptin signalling on insulin sensitivity and peripheral glucose metabolism. CONCLUSIONS/INTERPRETATION: This study represents the first line of genetic evidence that CRP is not merely a surrogate blood marker for inflammation and metabolic syndromes but directly regulates energy balance, body weight, insulin sensitivity and glucose homeostasis through direct regulation of leptin's central effect and hypothalamic signalling.


Assuntos
Proteína C-Reativa/genética , Resistência à Doença/genética , Resistência à Insulina/genética , Obesidade/genética , Animais , Peso Corporal/genética , Dieta Hiperlipídica , Metabolismo Energético/genética , Técnicas de Inativação de Genes , Masculino , Obesidade/metabolismo , Obesidade/patologia , Ratos , Ratos Transgênicos
5.
Gut ; 69(9): 1608-1619, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31900292

RESUMO

OBJECTIVE: High-fat diet (HFD)-induced metabolic disorders can lead to impaired sperm production. We aim to investigate if HFD-induced gut microbiota dysbiosis can functionally influence spermatogenesis and sperm motility. DESIGN: Faecal microbes derived from the HFD-fed or normal diet (ND)-fed male mice were transplanted to the mice maintained on ND. The gut microbes, sperm count and motility were analysed. Human faecal/semen/blood samples were collected to assess microbiota, sperm quality and endotoxin. RESULTS: Transplantation of the HFD gut microbes into the ND-maintained (HFD-FMT) mice resulted in a significant decrease in spermatogenesis and sperm motility, whereas similar transplantation with the microbes from the ND-fed mice failed to do so. Analysis of the microbiota showed a profound increase in genus Bacteroides and Prevotella, both of which likely contributed to the metabolic endotoxaemia in the HFD-FMT mice. Interestingly, the gut microbes from clinical subjects revealed a strong negative correlation between the abundance of Bacteroides-Prevotella and sperm motility, and a positive correlation between blood endotoxin and Bacteroides abundance. Transplantation with HFD microbes also led to intestinal infiltration of T cells and macrophages as well as a significant increase of pro-inflammatory cytokines in the epididymis, suggesting that epididymal inflammation have likely contributed to the impairment of sperm motility. RNA-sequencing revealed significant reduction in the expression of those genes involved in gamete meiosis and testicular mitochondrial functions in the HFD-FMT mice. CONCLUSION: We revealed an intimate linkage between HFD-induced microbiota dysbiosis and defect in spermatogenesis with elevated endotoxin, dysregulation of testicular gene expression and localised epididymal inflammation as the potential causes. TRIAL REGISTRATION NUMBER: NCT03634644.


Assuntos
Bacteroides/isolamento & purificação , Dieta Hiperlipídica/efeitos adversos , Disbiose , Prevotella/isolamento & purificação , Motilidade dos Espermatozoides/imunologia , Espermatogênese/imunologia , Animais , Correlação de Dados , Citocinas/análise , Disbiose/etiologia , Disbiose/microbiologia , Endotoxemia/microbiologia , Epididimo/imunologia , Epididimo/patologia , Fezes/microbiologia , Microbioma Gastrointestinal/imunologia , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Linfócitos T/imunologia
6.
Am J Pathol ; 188(7): 1597-1607, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29630856

RESUMO

Epithelial-to-mesenchymal transition (EMT) is postulated to be a prerequisite for the establishment of endometriosis (EMS), a common reproductive disorder in women. Our previous studies have demonstrated the elevated expression of transmembrane glycoprotein CD147 and its prosurvival effect on abnormal cells in endometriosis. Intriguingly, CD147 is known to promote EMT in cancers. However, the involvement of CD147 in EMT during the establishment of endometriosis remains incompletely understood. We found that CD147 promotes EMT in human endometrial adenocarcinoma cell line Ishikawa. We identified a novel CD147-interacting partner, cellular apoptosis susceptibility protein (CAS), which stabilized the interaction between E-cadherin (E-cad) and ß-catenin (ß-cat) by forming the CAS/E-cad/ß-cat complex. Down-regulation of CAS led to the release and nuclear translocation of ß-cat from E-cad, resulting in the overexpression of the EMT-promoting gene SNAIL. Interestingly, overexpression of CD147 impaired the interaction between CAS and E-cad and triggered the release of ß-cat from the CAS/E-cad/ß-cat complex, which in turn led to EMT. Furthermore, CAS was down-regulated in EMS, with elevated levels of CD147 and nuclear ß-cat. These findings suggest a previously undefined role of CAS in regulating EMT and reveal the involvement of a CD147-induced EMT signaling pathway in pathogenic progression of EMS.


Assuntos
Antígenos CD/metabolismo , Basigina/metabolismo , Caderinas/metabolismo , Proteína de Suscetibilidade a Apoptose Celular/metabolismo , Neoplasias do Endométrio/patologia , Endometriose/patologia , Transição Epitelial-Mesenquimal , beta Catenina/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adulto , Antígenos CD/genética , Basigina/genética , Caderinas/genética , Estudos de Casos e Controles , Membrana Celular , Proteína de Suscetibilidade a Apoptose Celular/genética , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Endometriose/genética , Endometriose/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais , Células Tumorais Cultivadas , beta Catenina/genética
7.
FASEB J ; 31(3): 1120-1129, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27974594

RESUMO

Perivascular adipose tissue (PVAT)-derived adiponectin (APN) is a secreted adipokine that protects against hypertension-related cardiovascular injury. However, the regulation of APN expression in hypertension remains to be explored. In this study, we demonstrated that down-regulation of APN was associated with complement activation in the PVAT of desoxycorticosterone acetate (DOCA)-salt hypertensive mice. Complement 3-deficient hypertensive mice were protected from ANP decrease in the PVAT. APN deficiency blockaded the protective effects of complement inhibition against hypertensive vascular injury. Mechanistically, complement 5a (C5a)-induced TNF-α secretion from macrophages is required for inhibiting APN expression in adipocytes. Macrophage depletion reversed C5a agonist peptide-induced TNF-α up-regulation and APN down-regulation in the PVAT of DOCA mice. Moreover, we detected increased macrophage infiltration and C5a expression associated with decreased APN expression in adipose tissue from patients with aldosterone-producing adenoma. These results identify a novel interaction between macrophages and adipocytes in the PVAT, where complement-mediated inhibition of APN acts as a potential risk factor for hypertensive vascular inflammation.-Ruan, C.-C., Ma, Y., Ge, Q., Li, Y., Zhu, L.-M., Zhang, Y., Kong, L.-R., Wu, Q-H., Li, F., Cheng, L., Zhao, A. Z., Zhu, D.-L., Gao, P.-J. Complement-mediated inhibition of adiponectin regulates perivascular inflammation and vascular injury in hypertension.


Assuntos
Adipócitos/metabolismo , Adiponectina/metabolismo , Complemento C3/metabolismo , Complemento C5a/metabolismo , Hipertensão/metabolismo , Remodelação Vascular , Adiponectina/genética , Animais , Regulação para Baixo , Humanos , Hipertensão/patologia , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
8.
Biochem J ; 473(9): 1215-24, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26933237

RESUMO

Defective central leptin signalling and impaired leptin entry into the CNS (central nervous system) represent two important aspects of leptin resistance in obesity. In the present study, we tested whether circulating human CRP (C-reactive protein) not only diminishes signalling of leptin within the CNS, but also impedes this adipokine's access to the CNS. Peripheral infusion of human CRP together with co-infused human leptin was associated with significantly decreased leptin content in the CSF of ob/ob mice. Furthermore, following peripheral infusion of human leptin, the CSF (cerebrospinal fluid) concentration of leptin in transgenic mice overexpressing human CRP was sharply lower than that achieved in similarly infused wild-type mice. Administration of LPS (lipopolysaccharide) to human CRP-transgenic mice dramatically elevated the concentrations of human CRP in the CSF. The i.c.v. (intracerebroventricular) delivery of human CRP into the lateral ventricles of ob/ob mice blocked the satiety and weight-reducing actions of human leptin, but not those of mouse leptin. I.c.v. injection of human CRP abolished hypothalamic signalling by human leptin, and ameliorated the effects of leptin on the expression of NPY (neuropeptide Y), AgRP (Agouti-related protein), POMC (pro-opiomelanocortin) and SOCS-3 (suppressor of cytokine signalling 3). Human CRP can impede the access of leptin to the CNS, and elevation of human CRP within the CNS can have a negative impact on the physiological actions of leptin.


Assuntos
Proteína C-Reativa , Hipotálamo/metabolismo , Leptina , Proteína Relacionada com Agouti/metabolismo , Animais , Proteína C-Reativa/farmacocinética , Proteína C-Reativa/farmacologia , Humanos , Leptina/farmacocinética , Leptina/farmacologia , Masculino , Camundongos , Camundongos Obesos , Neuropeptídeo Y/metabolismo , Pró-Opiomelanocortina/metabolismo , Transporte Proteico , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo
9.
Tumour Biol ; 37(9): 12241-12250, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27240591

RESUMO

S100A16 is a member of the S100 calcium-binding protein family. It is overexpressed in many types of tumors and associated with proliferation, migration, and invasion; however, its function in human prostate cancer is unresolved. Our objective was to determine its effects and the underlying pathways of S100A16 in prostate cancer tissues and cells. We measured S100A16 expression by quantitative real-time polymerase and Western blotting in eight matched prostate cancer and adjacent normal tissues, and in three prostate cancer cell lines, DU-145, LNCaP, and PC-3, compared to a normal prostate epithelial cell line PrEC. DU-145 cells stably overexpressing S100A16 and PC-3 cells with S100A16 knockdown were established by transfection with S100A16 overexpression plasmid or shRNAs. Invasion, migration, and proliferation were analyzed by transwell assay, wound healing, and colony formation assays, respectively. Western blotting and invasion assays were performed to determine expressions and activation of AKT, ERK, p21, and p27. S100A16 was significantly overexpressed in both prostate cancer tissues and cells lines compared to normal controls (P < 0.05). Overexpression of S100A16 significantly promoted invasion, migration, and proliferation in prostate cancer cells in vitro, whereas silencing S100A16 showed the converse effects (P < 0.05). Furthermore, overexpression of S100A16 activated cell signaling proteins AKT and ERK and downregulated tumor suppressors p21 and p27. Specific inhibitors, LY294002 and PD98059, suppressed activation of AKT and ERK, which attenuated DU-145 cell clone formation and invasion induced by S100A16 overexpression. S100A16 may promote human prostate cancer progression via signaling pathways involving AKT, ERK, p21, and p27 downstream effectors. Our findings suggest that S100A16 may serve as a novel therapeutic or diagnostic target in human prostate cancer.


Assuntos
Proliferação de Células , Sistema de Sinalização das MAP Quinases , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas S100/metabolismo , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Metástase Neoplásica , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas S100/genética
11.
Cell Physiol Biochem ; 36(4): 1670-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26160484

RESUMO

OBJECTIVE: This study evaluates the association between serum adiponectin concentrations and the risk of endometrial cancer through a comprehensive meta-analysis of currently available clinical data. METHODS: PubMed, Embase, the Chinese Biomedical Literature Database and the Science Citation Index (ISI Web of Science) were searched for studies that examined the association between blood adiponectin concentrations and the risk of endometrial cancer. Data from studies that met the inclusion criteria were systematically reviewed, and pooled analyses were performed according to the guidelines of Meta-Analysis of Observational Studies in Epidemiology and PRIMSA. RESULTS: Eight case-control studies (including 1257 endometrial cancer patients and 2008 controls) and four nested case-control studies (including 659 endometrial cancer patients and 1398 controls) were included. We found that serum adiponectin level was inversely correlated with the risk of endometrial cancer development after pooling the case-control studies (OR = 0.50, 95% CI: 0.39-0.60; P < 0.001). However, meta-analysis of nested case-control studies thus far did not support a broad linkage between serum adiponectin level and endometrial cancer, although a correlation may exist in the subgroup of postmenopausal women (OR=0.81, 95%CI: 0.65-1.00; P=0.060), particularly in postmenopausal women without current hormone replacement therapy (OR = 0.62, 95% CI: 0.44-0.86; P = 0.004). CONCLUSIONS: Meta-analysis of currently available clinical evidence supports the association between high serum adiponectin concentration and reduced risk of endometrial cancer development, particularly in the group of postmenopausal women without current hormone replacement therapy. However, additional studies with prospective design are required to fully support this linkage.


Assuntos
Adiponectina/sangue , Neoplasias do Endométrio/sangue , Neoplasias do Endométrio/epidemiologia , Endométrio/patologia , Neoplasias do Endométrio/patologia , Feminino , Humanos , Fatores de Risco
12.
Med Sci Monit ; 21: 1072-7, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25868851

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is the most common liver cancer, leading to many cancer-related deaths worldwide. Several studies have shown an association between pre-S deletion mutation of hepatitis B virus (HBV) and HCC risk, but the results remain conflicting. We aimed to verify HBV pre-S deletion mutations in relation to the risk of HCC. MATERIAL AND METHODS: We searched the commonly used electronic databases for relevant studies of this association among the Asian population until September 30th, 2014. Odds ratios (ORs) with 95% confidence intervals (CIs) were employed to calculate the association. RESULTS: A total of 17 case-control studies were screened out, including 2837 HBV-infected patients, of whom 1246 had HCC. The results showed that the frequency of pre-S deletion of HBV in patients with HCC was higher than that in patients without HCC (35.7% vs. 11.5%), indicating the prevalence of this mutation in patients with HCC. Statistically significant correlations were observed for pre-S deletion mutation and risk of HCC in a random-effects model (OR=3.90, 95% CI=2.80-5.44, P<0.00001). This association was also found in Chinese populations (OR=4.84, 95% CI=2.86-8.20, P<0.00001). CONCLUSIONS: Our data indicate that HBV pre-S deletion mutations might be associated with HCC risk. Their oncogenic role may be important in studying the potential mechanism of HBV hepatocarcinogenesis.


Assuntos
Carcinoma Hepatocelular/virologia , Predisposição Genética para Doença , Vírus da Hepatite B/genética , Neoplasias Hepáticas/virologia , Deleção de Sequência/genética , Adulto , Povo Asiático/genética , Estudos de Associação Genética , Humanos , Pessoa de Meia-Idade , Viés de Publicação , Fatores de Risco
13.
Int Immunopharmacol ; 138: 112659, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-38996665

RESUMO

Autoimmune hepatitis (AIH) is a chronic liver disease characterized by immune dysregulation and hepatocyte damage. FKBP38, a member of the immunophilin family, has been implicated in immune regulation and the modulation of intracellular signaling pathways; however, its role in AIH pathogenesis remains poorly understood. In this study, we aimed to investigate the effects of hepatic FKBP38 deletion on AIH using a hepatic FKBP38 knockout (LKO) mouse model created via cre-loxP technology. We compared the survival rates, incidence, and severity of AIH in LKO mice with those in control mice. Our findings revealed that hepatic FKBP38 deletion resulted in an unfavorable prognosis in LKO mice with AIH. Specifically, LKO mice exhibited heightened liver inflammation and extensive hepatocyte damage compared to control mice, with a significant decrease in anti-apoptotic proteins and a marked increase in pro-apoptotic proteins. Additionally, transcriptional and translational levels of pro-inflammatory cytokines and chemokines were significantly increased in LKO mice compared to control mice. Immunoblot analysis showed that MCP-1 expression was significantly elevated in LKO mice. Furthermore, the phosphorylation of p38 was increased in LKO mice with AIH, indicating that FKBP38 deletion promotes liver injury in AIH by upregulating p38 phosphorylation and increasing MCP-1 expression. Immune cell profiling demonstrated elevated populations of T, NK, and B cells, suggesting a dysregulated immune response in LKO mice with AIH. Overall, our findings suggest that FKBP38 disruption exacerbates AIH severity by augmenting the immune response by activating the MCP-1/p38 signaling pathway.


Assuntos
Quimiocina CCL2 , Hepatite Autoimune , Proteínas de Ligação a Tacrolimo , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Masculino , Camundongos , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Concanavalina A , Modelos Animais de Doenças , Hepatite Autoimune/imunologia , Fígado/patologia , Fígado/imunologia , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Transdução de Sinais , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo
14.
Cell Death Dis ; 15(5): 349, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769167

RESUMO

Osteosarcoma is a malignant bone tumor that primarily inflicts the youth. It often metastasizes to the lungs after chemotherapy failure, which eventually shortens patients' lives. Thus, there is a dire clinical need to develop a novel therapy to tackle osteosarcoma metastasis. Methionine dependence is a special metabolic characteristic of most malignant tumor cells that may offer a target pathway for such therapy. Herein, we demonstrated that methionine deficiency restricted the growth and metastasis of cultured human osteosarcoma cells. A genetically engineered Salmonella, SGN1, capable of overexpressing an L-methioninase and hydrolyzing methionine led to significant reduction of methionine and S-adenosyl-methionine (SAM) specifically in tumor tissues, drastically restricted the growth and metastasis in subcutaneous xenograft, orthotopic, and tail vein-injected metastatic models, and prolonged the survival of the model animals. SGN1 also sharply suppressed the growth of patient-derived organoid and xenograft. Methionine restriction in the osteosarcoma cells initiated severe mitochondrial dysfunction, as evident in the dysregulated gene expression of respiratory chains, increased mitochondrial ROS generation, reduced ATP production, decreased basal and maximum respiration, and damaged mitochondrial membrane potential. Transcriptomic and molecular analysis revealed the reduction of C1orf112 expression as a primary mechanism underlies methionine deprivation-initiated suppression on the growth and metastasis as well as mitochondrial functions. Collectively, our findings unraveled a molecular linkage between methionine restriction, mitochondrial function, and osteosarcoma growth and metastasis. A pharmacological agent, such as SGN1, that can achieve tumor specific deprivation of methionine may represent a promising modality against the metastasis of osteosarcoma and potentially other types of sarcomas as well.


Assuntos
Neoplasias Ósseas , Metionina , Mitocôndrias , Osteossarcoma , Osteossarcoma/patologia , Osteossarcoma/metabolismo , Osteossarcoma/genética , Osteossarcoma/tratamento farmacológico , Metionina/deficiência , Metionina/metabolismo , Humanos , Animais , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Linhagem Celular Tumoral , Camundongos , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Metástase Neoplásica , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/farmacologia , Camundongos Nus , Espécies Reativas de Oxigênio/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos
15.
Nat Med ; 12(4): 425-32, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16582918

RESUMO

The mechanisms underlying leptin resistance are still being defined. We report here the presence in human blood of several serum leptin-interacting proteins (SLIPs), isolated by leptin-affinity chromatography and identified by mass spectrometry and immunochemical analysis. We confirmed that one of the major SLIPs is C-reactive protein (CRP). In vitro, human CRP directly inhibits the binding of leptin to its receptors and blocks its ability to signal in cultured cells. In vivo, infusion of human CRP into ob/ob mice blocked the effects of leptin upon satiety and weight reduction. In mice that express a transgene encoding human CRP, the actions of human leptin were completely blunted. We also found that physiological concentrations of leptin can stimulate expression of CRP in human primary hepatocytes. Recently, human CRP has been correlated with increased adiposity and plasma leptin. Thus, our results suggest a potential mechanism contributing to leptin resistance, by which circulating CRP binds to leptin and attenuates its physiological functions.


Assuntos
Proteína C-Reativa/metabolismo , Leptina/metabolismo , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Proteína C-Reativa/farmacologia , Proteínas de Transporte/isolamento & purificação , Proteínas de Transporte/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Concentração Inibidora 50 , Interleucina-6/farmacologia , Leptina/sangue , Leptina/farmacologia , Camundongos , Camundongos Obesos , Camundongos Transgênicos , Testes de Precipitina , Proteínas de Ligação a RNA , Ratos , Transgenes
16.
BMC Med Genomics ; 16(1): 317, 2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-38057821

RESUMO

BACKGROUND: Glioma cells have increased intake and metabolism of methionine, which can be monitored with 11 C-L-methionine. However, a short half-life of 11 C (~ 20 min) limits its application in clinical practice. It is necessary to develop a methionine metabolism genes-based prediction model for a more convenient prediction of glioma survival. METHODS: We evaluated the patterns of 29 methionine metabolism genes in glioma from the Cancer Genome Atlas (TCGA). A risk model was established using Lasso regression analysis and Cox regression. The reliability of the prognostic model was validated in derivation and validation cohorts (Chinese Glioma Genome Atlas; CGGA). GO, KEGG, GSEA and ESTIMATE analyses were performed for biological functions and immune characterization. RESULTS: Our results showed that a majority of the methionine metabolism genes (25 genes) were involved in the overall survival of glioma (logrank p and Cox p < 0.05). A 7-methionine metabolism prognostic signature was significantly related to a poor clinical prognosis and overall survival of glioma patients (C-index = 0.83). Functional analysis revealed that the risk model was correlated with immune responses and with epithelial-mesenchymal transition. Furthermore, the nomogram integrating the signature of methionine metabolism genes manifested a strong prognostic ability in the training and validation groups. CONCLUSIONS: The current model had the potential to improve the understanding of methionine metabolism in gliomas and contributed to the development of precise treatment for glioma patients, showing a promising application in clinical practice.


Assuntos
Glioma , Humanos , Reprodutibilidade dos Testes , Prognóstico , Glioma/genética , Metionina , Racemetionina
17.
Biomed Pharmacother ; 157: 114027, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36436494

RESUMO

Acute myeloid leukemia (AML) is prone to drug-resistant relapse with a low 5-year survival rate. New therapeutic modalities are sorely needed to provide hope for AML relapse patients. Herein, we demonstrated a specific inhibitor of type 4 phosphodiesterase (PDE4), Zl-n-91, could significantly reduce the proliferation of AML cells, block DNA replication process, and increase AML cell death. Zl-n-91 also impeded the growth of subcutaneous xenograft and prolonged the survival of the MLL-AF9-driven AML model. Bioinformatic analysis revealed that elevated mitochondrial gene signatures inversely correlate with the survival of AML patients; and importantly, Zl-n-91 strongly suppressed the function of mitochondria. In addition, this PDE4 inhibitor induced alterations in multiple signaling pathways, including the reduction of ß-catenin activity. Stimulation of the Wnt/ß-catenin pathway could attenuate the inhibitory effect of Zl-n-91 on AML cell proliferation as well as mitochondrial function. Taken together, we revealed for the first time that targeting PDE4 activity could attenuate mitochondrial function through a Wnt/ß-catenin pathway, which in turn would block the growth of AML cells. Specific PDE4 inhibitors can potentially serve as a new treatment modality for AML patients.


Assuntos
Leucemia Mieloide Aguda , beta Catenina , Humanos , beta Catenina/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Via de Sinalização Wnt , Leucemia Mieloide Aguda/metabolismo , Proliferação de Células , Mitocôndrias/metabolismo , Linhagem Celular Tumoral
18.
Cell Rep Med ; 4(6): 101070, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37269826

RESUMO

The strong dependency of almost all malignant tumors on methionine potentially offers a pathway for cancer treatment. We engineer an attenuated strain of Salmonella typhimurium to overexpress an L-methioninase with the aim of specifically depriving tumor tissues of methionine. The engineered microbes target solid tumors and induce a sharp regression in several very divergent animal models of human carcinomas, cause a significant decrease in tumor cell invasion, and essentially eliminate the growth and metastasis of these tumors. RNA sequencing analyses reveal that the engineered Salmonella reduce the expression of a series of genes promoting cell growth, cell migration, and invasion. These findings point to a potential treatment modality for many metastatic solid tumors, which warrants further tests in clinical trials.


Assuntos
Metionina , Neoplasias , Animais , Humanos , Metionina/metabolismo , Metionina/uso terapêutico , Neoplasias/tratamento farmacológico , Racemetionina/metabolismo , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Modelos Animais
19.
Cancers (Basel) ; 14(23)2022 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-36497484

RESUMO

NPC is a type of malignant tumor with a high risk of local invasion and early distant metastasis. Resistin is an inflammatory cytokine that is predominantly produced from the immunocytes in humans. Accumulating evidence has suggested a clinical association of circulating resistin with the risk of tumorigenesis and a relationship between blood resistin levels and the risk of cancer metastasis. In this study, we explored the blood levels and the role of resistin in NPC. High resistin levels in NPC patients were positively associated with lymph node metastasis, and resistin promoted the migration and invasion of NPC cells in vitro. These findings were also replicated in a mouse model of NPC tumor metastasis. We identified TLR4 as a functional receptor in mediating the pro-migratory effects of resistin in NPC cells. Furthermore, p38 MAPK and NF-κB were intracellular effectors that mediated resistin-induced EMT. Taken together, our results suggest that resistin promotes NPC metastasis by activating the TLR4/p38 MAPK/NF-κB signaling pathways.

20.
Med Oncol ; 37(5): 43, 2020 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-32270348

RESUMO

Breast cancer is the most common cancer in women. Although several studies demonstrated cellular apoptosis susceptibility protein (CAS) involved in the development of breast cancer, the underlying mechanisms of CAS regulating cell processes in the breast cancer remain elusive. In the present study, we explored the possible mechanism of CAS in contributing to the cell proliferation in the breast cancer cell line MCF-7. Knockdown of CAS led to the reduction of cell viability and proliferation. Furthermore, cell cycle was arrested in G0/G1 phase after knocking down CAS with the decrease of cyclinD1. In addition, RNA-seq analysis for the CAS knockdown cells demonstrated that total eleven genes were significantly altered (Fold changes > 2). Of note, the expression of cyp24a1 was dramatically increased in the shCAS cells compared to that of shNC cells as well as confirmed by quantitative real-time polymerase chain reaction (qPCR). These observations clarified the previous conflicting results on the cell fates of the breast cells regulated by CAS and provide new insight into the role of CAS in the development of breast cancer.


Assuntos
Neoplasias da Mama/patologia , Proliferação de Células/genética , Proteína de Suscetibilidade a Apoptose Celular/metabolismo , Vitamina D3 24-Hidroxilase/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Ciclo Celular/genética , Sobrevivência Celular/genética , Proteína de Suscetibilidade a Apoptose Celular/genética , Bases de Dados Genéticas , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA