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












Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 30(7): 1742-1756, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37188742

RESUMEN

In the past few decades, advances in the outcomes of patients suffering from pancreatic ductal adenocarcinoma (PDAC) have lagged behind these gained in the treatment of many other malignancies. Although the pivotal role of the SUMO pathway in PDAC has been illustrated, the underlying molecule drivers have yet to be fully elucidated. In the present study, we identified SENP3 as a potential suppressor of PDAC progression through an in vivo metastatic model. Further studies revealed that SENP3 inhibited PDAC invasion in a SUMO system dependent fashion. Mechanistically, SENP3 interacted with DKC1 and, as such, catalyzed the deSUMOylation of DKC1, which accepted SUMO3 modifiers at three lysine residues. SENP3-mediated deSUMOylation caused DKC1 instability and disruption of the interaction between snoRNP proteins, which contributed to the impaired migration ability of PDAC. Indeed, overexpression of DKC1 abated the anti-metastasis effect of SENP3, and DKC1 was elevated in PDAC specimens and associated with a poor prognosis in PDAC patients. Collectively, our findings shed light on the essential role of SENP3/DKC1 axis in the progression of PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Movimiento Celular , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Péptido Hidrolasas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proliferación Celular , Proteínas Nucleares/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Neoplasias Pancreáticas
2.
Clin Transl Med ; 13(3): e1213, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36855786

RESUMEN

BACKGROUND: Intrahepatic cholangiocarcinoma (iCCA) is a highly lethal malignancy characterized by massive fibrosis and has ineffective adjuvant therapies. Here, we demonstrate the potential of angiotensin receptor blockers (ARBs) in targeting iCCA. METHODS: Masson's trichrome staining was used to assess the effect of ARBs in iCCA specimens, CCK8 and gel contraction assays in vitro and in xenograft models in vivo. RNA-seq and ATAC-seq were used for mechanistic investigations. RESULTS: Patients with iCCA who were administered ARBs had a better prognosis and a lower proportion of tumour stroma, indicating alleviated fibrosis. The presence of AGTR1, the ARBs receptor, is associated with a poor prognosis of iCCA and is highly expressed in tumour tissues and cancer-associated fibroblasts (CAFs). The ARBs strongly attenuated the viability of AGTR1+ CAFs in vitro and retarded tumour progression and fibrosis in xenograft models of co-cultured CAFs and iCCA cells. Still, they did not have a significant effect on AGTR1- CAFs. Moreover, ARBs decreased the secretion of AGTR1+ CAF-derived MFAP5 via the Hippo pathway, weakened the interaction between CAFs and iCCA cells, and impaired the aggressiveness of iCCA cells by attenuating the activation of the Notch1 pathway in iCCA cells. CONCLUSIONS: ARBs exhibit anti-fibrotic function by inhibiting the viability of AGTR1+ CAFs. These findings support using ARBs as a novel therapeutic option for targeting iCCA.


Asunto(s)
Neoplasias de los Conductos Biliares , Fibroblastos Asociados al Cáncer , Colangiocarcinoma , Animales , Humanos , Antagonistas de Receptores de Angiotensina/farmacología , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina , Colangiocarcinoma/tratamiento farmacológico , Modelos Animales de Enfermedad , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Conductos Biliares Intrahepáticos , Receptor de Angiotensina Tipo 1/genética
3.
Cancer Sci ; 113(11): 3735-3750, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36047966

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal malignancy with extremely poor prognosis. Gemcitabine resistance is a major challenge in the treatment of PDAC. Here, we showed that LINC00460 was associated with the response to gemcitabine both in PDAC patients and PDAC-PDX. After knocking down LINC00460 in PDAC tumor cells, results of RNA sequencing followed by gene ontology analysis indicated that LINC00460 influenced the activity of growth factors and modified the extracellular matrix. FISH showed that LINC00460 is mostly located in the cytoplasm. Results of RNA pull-down, LC-MS/MS, RIP, and immunoblotting confirmed that LINC00460 could directly bind to PDAP1. Furthermore, we demonstrated that LINC00460 mediated the cellular communication of PDAC tumor cells and CAFs by PDAP1/PDGFA/PDGFR signaling pathway and regulated the gemcitabine-resistance function of CAFs, which could be reversed by treatment with a PDGFR inhibitor (crenolanib). PDAC-PDX tumors with lower expression of LINC00460 showed a better response to gemcitabine plus crenolanib treatment. Our finding supported the application of LINC00460 in precision medicine that uses gemcitabine plus crenolanib to treat PDAC with low expression of LINC00460.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , ARN Largo no Codificante , Humanos , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Cromatografía Liquida , Resistencia a Antineoplásicos/genética , Péptidos y Proteínas de Señalización Intercelular , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Espectrometría de Masas en Tándem , ARN Largo no Codificante/genética , Gemcitabina , Neoplasias Pancreáticas
4.
Clin Transl Med ; 12(6): e848, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35696608

RESUMEN

BACKGROUND: Intrahepatic cholangiocarcinoma (ICC) is an aggressive cancer with exceedingly poor prognosis, and chemoresistance is a huge challenge for treatment. N6-methyladenosine (m6 A) modification plays an important role in the progression and chemoresistance of cancers. We aimed to investigate the oncogenic function and therapeutic significance of the m6 A binding protein, YTH domain family 2 (YTHDF2), in ICC progression and cisplatin-based chemotherapy. METHODS: Several independent data sets were used to assess the expression of YTHDF2 in ICC, particularly in chemoresistant ICC. Knockdown and overexpression were used to evaluate the effects of YTHDF2 on tumourigenesis and cisplatin response in ICC. Multi-omics sequencing was performed to identify target genes. RIP, dual luciferase reporter, RNA stability experiment and loss-of-function assays were conducted to study the mechanisms underlying the oncogenic function of YTHDF2. Furthermore, patient-derived xenograft (PDX) model was established to determine the effect of combination treatment of YTHDF2 siRNA and cisplatin in ICC. RESULTS: Our study showed that YTHDF2 was upregulated in ICC tissues, particularly in chemoresistant ICC tissues, and correlated with poor prognosis. Furthermore, silencing YTHDF2 led to inhibited proliferation, promoted apoptosis and G0/G1 cell cycle arrest. Its downregulation also enhanced DNA damage and sensitised ICC cells to cisplatin. YTHDF2 overexpression exerted the opposite results. Integration analysis using RNA-seq, MeRIP-seq and anti-YTHDF2 RIP-seq elucidated the role of YTHDF2 in tumourigenesis and cisplatin-desensitising function by promoting the degradation of cyclin-dependent kinase inhibitor 1B (CDKN1B) mRNA in an m6 A-dependent manner. Downregulation of CDKN1B increased the YTHDF2 silencing-induced influence on tumourigenesis and cisplatin response to ICC. In addition, the combination treatment of YTHDF2 siRNA and cisplatin significantly enhanced the anti-tumour effect of cisplatin in a chemoresistant ICC PDX model. CONCLUSIONS: YTHDF2 exhibits tumour oncogenic and cisplatin-desensitising properties, which may offer insight into the development of novel combination therapeutic strategies for ICC.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular/genética , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Cisplatino/metabolismo , Cisplatino/farmacología , Cisplatino/uso terapéutico , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/uso terapéutico , Humanos , Estabilidad del ARN/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Proteínas de Unión al ARN/genética
5.
Cancer Lett ; 544: 215802, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-35732215

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with poor prognosis. Its fibrotic tumor microenvironment (TME) plays a crucial role in promoting tumor invasion and metastasis, which eventually leads to a dismal 5-year survival rate in PDAC patients. Aortic carboxypeptidase-like protein (ACLP) promotes tissue fibrosis in benign diseases. However, its role in cancer-associated fibrosis remains unelucidated. Here, we show that ACLP was mainly expressed in cancer-associated fibroblasts (CAFs) but not in cancer cells and highly expressed in PDAC tissues. High ACLP expression was correlated with poor overall survival. Moreover, ACLP expression in PDAC patients with liver metastases was higher than that in PDAC patients without liver metastases. By detecting activation marker expression and CAF contractility and motility, we found that ACLP promoted CAF activation in PDAC, leading to TME fibrosis. Furthermore, ACLP-activated CAFs could promote cancer cell invasion in vitro and tumor metastasis in vivo. Mechanistically, ACLP promotes the expressions of MMP1 and MMP3 in CAFs, thus promoting PDAC invasion and metastasis. Intriguingly, we identified an ACLP-PPARγ-ACLP feedback loop in PDAC CAFs. Abatement of this feedback loop might be a promising approach in CAF-targeting PDAC treatment.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carboxipeptidasas/metabolismo , Carcinoma Ductal Pancreático , Neoplasias Hepáticas , Neoplasias Pancreáticas , Proteínas Represoras/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Retroalimentación , Fibrosis , Humanos , Neoplasias Hepáticas/patología , PPAR gamma/genética , PPAR gamma/metabolismo , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Neoplasias Pancreáticas
6.
Front Immunol ; 13: 773291, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35265067

RESUMEN

Neutrophil infiltration plays an important role in the initial phase of hepatic ischemia and reperfusion injury (HIRI). Despite many different key molecules that have been reported to meditate neutrophil trafficking in HIRI, the mechanism of this process has not been fully elucidated. In this study, we found that Carabin deficiency in myeloid cells (LysMCre : Carabinfl/fl) aggravated IRI-induced hepatic injury and apoptosis through increasing the infiltration of CD11b+Ly6G+ neutrophils. ImmGen Datasets further revealed that Carabin was expressed in bone marrow neutrophils (GM.BM) but was significantly downregulated in thio-induced peripheral neutrophils (GN.Thio.PC), which was consistently verified by comparing GM.BM and liver-infiltrating neutrophils induced by IRI. Mechanistically, up-regulation of Carabin in GM.BM in vitro reduced the expression levels of P-selectin, E-selectin, and αvß3 integrin through inhibiting Ras-ERK and Calcineurin-NFAT signaling. Furthermore, blocking P-selectin, E-selectin, and αvß3 integrin in LysMCre : Carabinfl/fl mice decreased the frequency and number of CD11b+Ly6G+ neutrophils and reversed hepatic ischemia-reperfusion damage. In conclusion, our results provide a new understanding of Carabin, such that it is expressed and functions not only in adaptive immune cells (T and B cells) but also in innate immune cells (neutrophils), contributing to the migration of neutrophils. These findings provide novel and promising therapeutic targets for the prevention of HIRI during liver transplantation or hepatic surgery.


Asunto(s)
Neutrófilos , Daño por Reperfusión , Animales , Calcineurina/metabolismo , Selectina E/metabolismo , Integrinas/metabolismo , Isquemia/metabolismo , Hígado/metabolismo , Ratones , Neutrófilos/metabolismo , Daño por Reperfusión/metabolismo
7.
Front Immunol ; 12: 535012, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093514

RESUMEN

Rejection after organ transplantation is a cause of graft failure. Effectively reducing rejection and inducing tolerance is a challenge in the field of transplantation immunology. The liver, as an immunologically privileged organ, has high rates of spontaneous and operational tolerance after transplantation, allowing it to maintain its normal function for long periods. Although modern immunosuppression regimens have serious toxicity and side effects, it is very risky to discontinue immunosuppression regimens blindly. A more effective treatment to induce immune tolerance is the most sought-after goal in transplant medicine. Tregs have been shown to play a pivotal role in the regulation of immune balance, and infusion of Tregs can also effectively prevent rejection and cure autoimmune diseases without significant side effects. Given the immune characteristics of the liver, the correct use of Tregs can more effectively induce the occurrence of operational tolerance for liver transplants than for other organ transplants. This review mainly summarizes the latest research advances regarding the characteristics of the hepatic immune microenvironment, operational tolerance, Treg generation in vitro, and the application of Tregs in liver transplantation. It is hoped that this review will provide a deeper understanding of Tregs as the most effective treatment to induce and maintain operational tolerance after liver transplantation.


Asunto(s)
Investigación Biomédica/métodos , Tolerancia Inmunológica/inmunología , Trasplante de Hígado/métodos , Linfocitos T Reguladores/inmunología , Inmunología del Trasplante/inmunología , Tolerancia al Trasplante/inmunología , Investigación Biomédica/tendencias , Predicción , Rechazo de Injerto/inmunología , Humanos , Hígado/inmunología
8.
Front Genet ; 11: 577040, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33304383

RESUMEN

OBJECTIVE: A challenging issue in the clinical management of lupus nephritis (LN) is the resistance to immunosuppressive therapy. We postulated that perturbed intrarenal immune cell landscape affected LN onset and remission induction, and shedding light on the characteristics of intrarenal immune cell infiltration could cultivate more efficient treatment regimens. MATERIALS AND METHODS: Genome-wide expression profiles of microarray datasets were downloaded from the Gene Expression Omnibus database. The CIBERSORT algorithm was used to analyze the intrarenal immune cell landscape, followed by Pearson correlation analysis and principal component analysis. The differentially expressed genes were identified and subjected to Gene Ontology (GO) enrichment analyses and protein-protein interaction network establishment, being visualized by Cytoscape and further analyzed by CytoHubba to extract hub genes. Hub genes were also validated in the genomic dataset from kidney biopsy-proven LN patients. RESULTS: In addition to memory B cells, monocytes and M1 macrophages were identified as two predominantly increased intrarenal immune cell types in LN-prone NZB/W mice upon nephritis onset. Most interestingly, apart from memory B cells, monocytes and M1 macrophages proportions in kidney tissue were significantly lower in early remission mice compared with late remission mice. Furthermore, GO analysis showed that intrarenal mononuclear phagocytes triggered nephritis onset mainly via the initiation of adaptive immune response and inflammatory reaction, but this functional involvement was mitigated upon remission induction. Hub genes related to LN onset in NZB/W mice were validated in the genomic dataset from kidney biopsy-proven LN patients. CONCLUSION: LN characterizes aberrant mononuclear phagocytes abundance and signature upon disease onset, of which the reversal is associated with early remission induction in LN-prone NZB/W mice. Mononuclear phagocytes might be an adjunctive histology marker for monitoring disease onset and stratifying LN patients in terms of response to remission induction therapy.

9.
Ann Transl Med ; 8(14): 856, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32793700

RESUMEN

BACKGROUND: Cancer-associated fibroblasts (CAFs) are a major component of hepatocellular carcinoma (HCC) stroma that are critically involved in HCC cancer chemoresistance, but the mechanism has not been elucidated. Previous studies have reported CD73 exerted an immunosuppressive function in cancer. Here, we explored the mechanism by which CAFs regulates CD73+ HCC cells and clarified whether CAFs promote chemoresistance of CD73+ cells. METHODS: We used the co-culture method to study the relationship between CAFs and HCC cells. Immunohistochemistry was applied to evaluate the correlation between α-smooth-muscle actin (α-SMA) and CD73. CD73 mRNA and protein were determined by real-time polymerase chain reaction (RT-PCR) and western blotting, and hepatocyte growth factor (HGF) was assayed by enzyme-linked immunosorbent assay (ELISA). Western blotting was used to explore the regulated pathway of CD73+ HCC. We then knocked down CD73 in cells, and then assessed the effect of CD73 on the apoptosis by flow cytometry. Finally, a sphere formation assay was applied to investigate the stemness of cancer cells, and xenograft tumors in nude mice were built to investigate the tumorigenicity. RESULTS: We found that the proportion of CAFs was positively correlated with CD73 expression in HCC cells. Mechanistically, c-Met and the MEK-ERK1/2 pathway were activated by HGF from CAFs which upregulated CD73 expression in HCC cells. Also, we found that CD73 promote sorafenib and cisplatin resistance in HCC, and CD73+ HCC cells indicated the higher capability of tumorigenicity compared to CD73- HCC cells in vivo. Furthermore, HGF further enhanced the chemoresistant characteristics of CD73+ tumor cells. CONCLUSIONS: Our findings collectively suggest that CD73 is a vital HCC-chemoresistance force controlled by cross-talking between CAFs and HCC cells, thereby establishing CD73 as a potential new therapeutic target for HCC.

10.
Ann Transl Med ; 7(18): 465, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31700901

RESUMEN

BACKGROUND: Regulatory B cells (Bregs) play an essential role in inflammation and transplant tolerance. Several studies have reported a decreased number of Bregs in renal transplant patients with graft rejection. However, little is known about their role in the liver alloresponse. METHODS: To investigate whether the circulating Bregs have been associated with acute allograft rejection (AR) in liver transplantation patients, 19 patients receiving liver allografts from donation after cardiac death (DCD) donors were retrospectively studied. RESULTS: The postoperative proportions of circulating CD19+CD24hiCD38hi transitional Bregs (tBregs) and CD19+CD24hiCD27+ memory Bregs (mBregs) in patients diagnosed with AR (AR group) and other patients with stable allograft liver function (SF group) were evaluated using flow cytometry (FCM) analysis. Results showed that while no significant changes were found regarding both the tBreg and mBreg, proportions across all time points in the SF group, the AR group showed significantly decreased proportions of mBregs. All of the five AR patients responded fine to the treatments, and the proportions of mBregs increased significantly after anti-rejection therapies. In addition, AR was suspected in four recipients, but gradually they were diagnosed with hemolytic or obstructive jaundice and showed no decrease in the proportion of mBregs. CONCLUSIONS: For the first time, our results suggested the potential role of a decreased proportion of circulating mBregs in predicting AR in patients with post liver transplantation.

11.
Int Immunopharmacol ; 77: 105965, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31670092

RESUMEN

Tumors escape immune attacks via various mechanisms, among which activation of regulatory pathways in effector immune cells and recruitment of immunosuppressive cells are usually employed. Traf6 is a member of the family of tumor necrosis factor receptor-associated factors and involved in many signaling pathways. While it plays important roles in both tumor biology and immune system, the potential therapeutic role of Traf6 in tumor immunotherapy hasn't ever been assessed. Here, we confirmed the anti-tumor effect of Traf6 inhibitor in Hepa1-6 tumor model. Flow cytometry-based analysis revealed that T cell-mediated antitumor immunity was provoked and the infiltration of Treg cells was restrained when treated with Traf6 inhibitor. Via an in vivo migration assay, we found that Traf6 inhibitor decreased the population of intratumor Tregs by impeding the migration of Tregs towards tumor. Finally, we demonstrated that combination of Traf6 inhibitor and PD-1 blockade could receive a better antitumor efficiency. These results implicated that Traf6 inhibitor could serve as a supplement for immune checkpoint therapy.


Asunto(s)
Movimiento Celular/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología , Animales , Línea Celular Tumoral , Femenino , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL
12.
EMBO J ; 38(9)2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30886050

RESUMEN

Regulatory T cells (Tregs) are crucial mediators of immune control. The characteristic gene expression and suppressive functions of Tregs depend considerably on the stable expression and activity of the transcription factor FOXP3. Transcriptional regulation of the Foxp3 gene has been studied in depth, but both the expression and function of this factor are also modulated at the protein level. However, the molecular players involved in posttranslational FOXP3 regulation are just beginning to be elucidated. Here, we found that TRAF6-deficient Tregs were dysfunctional in vivo; mice with Treg-restricted deletion of TRAF6 were resistant to implanted tumors and displayed enhanced anti-tumor immunity. We further determined that FOXP3 undergoes K63-linked ubiquitination at lysine 262 mediated by the E3 ligase TRAF6. In the absence of TRAF6 activity or upon mutation of the ubiquitination site, FOXP3 displayed aberrant, perinuclear accumulation and disrupted regulatory function. Thus, K63-linked ubiquitination by TRAF6 ensures proper localization of FOXP3 and facilitates the transcription factor's gene-regulating activity in Tregs. These results implicate TRAF6 as a key posttranslational, Treg-stabilizing regulator that may be targeted in novel tolerance-breaking therapies.


Asunto(s)
Colitis/inmunología , Factores de Transcripción Forkhead/fisiología , Lisina/metabolismo , Melanoma Experimental/inmunología , Linfocitos T Reguladores/inmunología , Factor 6 Asociado a Receptor de TNF/fisiología , Ubiquitinación , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
13.
Cell Death Dis ; 10(1): 13, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622237

RESUMEN

Human gingival tissue-derived mesenchymal stem cells (GMSCs) present an accessible source of mesenchymal stem cells (MSCs) for treating autoimmune diseases. Here we show that human GMSCs can prevent and treat acute graft-versus-host disease (GVHD) in two different mouse models. Our results indicate that besides exhibiting suppressive function in vitro and in vivo, GMSCs may also regulate the conversion of Tregs to Th1 and/or Th17-like cells, as well as stabilize Foxp3 expression. Furthermore, GMSC-mediated prevention of acute GVHD was dependent on CD39 signaling that play an important role in the function and stability of Tregs. Finally, we also observed stronger protective ability of GMSCs with greater expansion ability compared with BMSCs or ASCs. These results indicate that human GMSCs have the potential to be used to treat GVHD.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Encía/citología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Mesenquimatosas , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Células TH1/inmunología , Células Th17/inmunología
14.
Cell Death Dis ; 9(9): 910, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30185770

RESUMEN

Activating transcription factor 3 (ATF3) is a stress-induced transcription factor that plays important roles in regulating immune and metabolic homeostasis. Activation of the mechanistic target of rapamycin (mTOR) and hypoxia-inducible factor (HIF) transcription factors are crucial for the regulation of immune cell function. Here, we investigated the mechanism by which the ATF3/mTOR/HIF-1 axis regulates immune responses in a liver ischemia/reperfusion injury (IRI) model. Deletion of ATF3 exacerbated liver damage, as evidenced by increased levels of serum ALT, intrahepatic macrophage/neutrophil trafficking, hepatocellular apoptosis, and the upregulation of pro-inflammatory mediators. ATF3 deficiency promoted mTOR and p70S6K phosphorylation, activated high mobility group box 1 (HMGB1) and TLR4, inhibited prolyl-hydroxylase 1 (PHD1), and increased HIF-1α activity, leading to Foxp3 downregulation and RORγt and IL-17A upregulation in IRI livers. Blocking mTOR or p70S6K in ATF3 knockout (KO) mice or bone marrow-derived macrophages (BMMs) downregulated HMGB1, TLR4, and HIF-1α and upregulated PHD1, increasing Foxp3 and decreasing IL-17A levels in vitro. Silencing of HIF-1α in ATF3 KO mice ameliorated IRI-induced liver damage in parallel with the downregulation of IL-17A in ATF3-deficient mice. These findings demonstrated that ATF3 deficiency activated mTOR/p70S6K/HIF-1α signaling, which was crucial for the modulation of TLR4-driven inflammatory responses and T cell development. The present study provides potential therapeutic targets for the treatment of liver IRI followed by liver transplantation.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/metabolismo , Hepatopatías/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/fisiología , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Hepatopatías/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Neutrófilos/patología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Regulación hacia Arriba/fisiología
15.
Cancer Discov ; 8(8): 1026-1043, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29907586

RESUMEN

Regulatory T cells (Treg) are critical for maintaining self-tolerance and immune homeostasis, but their suppressive function can impede effective antitumor immune responses. FOXP3 is a transcription factor expressed in Tregs that is required for their function. However, the pathways and microenvironmental cues governing FOXP3 expression and Treg function are not completely understood. Herein, we report that YAP, a coactivator of the Hippo pathway, is highly expressed in Tregs and bolsters FOXP3 expression and Treg function in vitro and in vivo. This potentiation stemmed from YAP-dependent upregulation of activin signaling, which amplifies TGFß/SMAD activation in Tregs. YAP deficiency resulted in dysfunctional Tregs unable to suppress antitumor immunity or promote tumor growth in mice. Chemical YAP antagonism and knockout or blockade of the YAP-regulated activin receptor similarly improved antitumor immunity. Thus, we identify YAP as an unexpected amplifier of a Treg-reinforcing pathway with significant potential as an anticancer immunotherapeutic target.Significance: Tregs suppress antitumor immunity, and pathways supporting their function can be novel immunotherapy targets. Here, the selective expression of YAP by Tregs, its importance for their function, and its unexpected enhancement of pro-Treg Activin/SMAD signaling are reported, as are validations of potential cancer-fighting antagonists of YAP and its regulatory targets. Cancer Discov; 8(8); 1026-43. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 899.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción Forkhead/metabolismo , Neoplasias Experimentales/patología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Linfocitos T Reguladores/inmunología , Activinas/metabolismo , Adulto , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Transducción de Señal , Factores de Transcripción , Microambiente Tumoral , Regulación hacia Arriba , Proteínas Señalizadoras YAP
16.
Cell Physiol Biochem ; 46(6): 2261-2270, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29734164

RESUMEN

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and has the third highest mortality rate among all cancers. MicroRNAs are a class of endogenous, single-stranded short noncoding RNAs. The purpose of this study was to study the role of microRNA-873 in HCC. METHODS: The expression of miRNA-873 and tumor suppressor in lung cancer 1 (TSLC1) in HCC tissues and cell lines was detected by real-time quantitative RT-PCR (RT-qPCR) or western blot. A CCK-8 assay was used to examine cell proliferation; flow cytometry was used to assess the cell cycle; the Transwell migration assay was used to test for metastasis. Luciferase assays were performed to assess whether TSLC1 was a novel target of miRNA-873. RESULTS: We showed that miRNA-873 was upregulated in HCC tissues and cell lines compared with the normal control. Knockdown of miRNA-873 inhibited the growth and metastasis of HepG2 and accelerated G1 phase arrest, while overexpression of miRNA-873 had the opposite effect. The dual-luciferase reporter assays revealed that TSLC1 was a novel target of miRNA-873. Further study showed that TSLC1 was decreased in HCC tissues and cell lines. There was a negative correlation between the expression levels of TSLC1 and miRNA-873. The effect of miRNA-873 overexpression was neutralized by TSLC1. We also found that miRNA-873 activated the PI3K/AKT/mTOR signaling pathway and promoted HCC. CONCLUSIONS: Our data demonstrated that miRNA-873 promoted HCC progression by targeting TSLC1 and provided a new target for the therapy of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Molécula 1 de Adhesión Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , MicroARNs/genética , Invasividad Neoplásica/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/patología
17.
Proc Natl Acad Sci U S A ; 115(6): E1239-E1248, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29367423

RESUMEN

Triple-negative breast cancer (TNBC) is treated with cytotoxic chemotherapy and is often characterized by early relapse and metastasis. To form a secondary (recurrent and/or metastatic) tumor, a breast cancer cell must evade the innate and adaptive immune systems. CD47 enables cancer cells to evade killing by macrophages, whereas CD73 and PDL1 mediate independent mechanisms of evasion of cytotoxic T lymphocytes. Here, we report that treatment of human or murine TNBC cells with carboplatin, doxorubicin, gemcitabine, or paclitaxel induces the coordinate transcriptional induction of CD47, CD73, and PDL1 mRNA and protein expression, leading to a marked increase in the percentage of CD47+CD73+PDL1+ breast cancer cells. Genetic or pharmacological inhibition of hypoxia-inducible factors (HIFs) blocked chemotherapy-induced enrichment of CD47+CD73+PDL1+ TNBC cells, which were also enriched in the absence of chemotherapy by incubation under hypoxic conditions, leading to T cell anergy or death. Treatment of mice with cytotoxic chemotherapy markedly increased the intratumoral ratio of regulatory/effector T cells, an effect that was abrogated by HIF inhibition. Our results delineate an HIF-dependent transcriptional mechanism contributing to TNBC progression and suggest that combining chemotherapy with an HIF inhibitor may prevent countertherapeutic induction of proteins that mediate evasion of innate and adaptive antitumor immunity.


Asunto(s)
5'-Nucleotidasa/metabolismo , Antineoplásicos/farmacología , Antígeno B7-H1/metabolismo , Antígeno CD47/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias de la Mama Triple Negativas/inmunología , Escape del Tumor/inmunología , 5'-Nucleotidasa/genética , Animales , Apoptosis , Antígeno B7-H1/genética , Antígeno CD47/genética , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cell Mol Immunol ; 14(6): 521-528, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27374793

RESUMEN

Autoimmune diseases are characterized by an imbalance between regulatory T cells and effector T-cell subsets, such as Th1 and Th17 cells. Studies have confirmed that natural CD4+Foxp3+ Tregs were unstable and dysfunctional in the presence of pro-inflammatory cytokines. In the current study, human CD39hi Tregs and CD39low Tregs were sorted from Tregs in vitro after 7 days of expansion. The functions of both Treg subsets were investigated under inflammatory conditions in vitro and in vivo. In the presence of IL-1ß and IL-6, cultured CD4+CD39hi Tregs maintained stable forkhead box protein 3 expression, whereas CD4+CD39low Tregs lost Foxp3 expression and trans-differentiated into Th1 or Th17 cells. Decreased IL-1ßR and IL-6R expression on the CD39hi Tregs was the primary mechanism responsible for Treg stability. In addition, reduced activation of downstream molecules, such as STAT1 and STAT3, through the modulation of CpG demethylation played an important role. Finally, human CD4+CD39hi Tregs but not CD4+CD39low Tregs protected against xenograft versus host disease in model mice. These results strongly implied the physiological importance of CD39 expression and suggested that manipulation of CD39hi Tregs might represent a novel strategy for the treatment of autoimmune diseases.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Inflamación/inmunología , Trasplante de Células Madre , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Autoinmunidad , Células Cultivadas , Metilación de ADN , Factores de Transcripción Forkhead/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones SCID , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Trasplante Heterólogo
19.
Blood ; 128(10): 1424-35, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27485827

RESUMEN

CD4(+)CD25(+)FoxP3(+) thymic-derived regulatory T cells (tTregs) are indispensable for maintaining immune system equilibrium. Adoptive transfer of tTregs is an effective means of suppressing graft-versus-host disease (GVHD) in murine models and in early human clinical trials. Tumor necrosis factor receptor-associated factor 6 (TRAF6), an ubiquitin-conjugating enzyme that mediates nuclear factor κB (NF-κB) activation, plays an essential role in modulating regulatory T cell survival and function. MicroRNAs (miRNAs) are noncoding RNAs, which mediate RNA silencing and posttranscriptional gene repression. By performing comprehensive TaqMan Low Density Array miRNA assays, we identified 10 miRNAs differentially regulated in human tTreg compared with control T cells. One candidate, miR-146b, is preferentially and highly expressed in human naive tTregs compared with naive CD4 T cells. miRNA prediction software revealed that TRAF6 was the one of the top 10 scored mRNAs involved tTreg function with the highest probability as a potential miR-146b target. Antagomir-mediated knockdown of miRNA-146b, but not another miRNA-146 family member (miRNA-146a), enhanced TRAF6 expression. TRAF6, in turn, increases NF-κB activation, which is essential for tTreg function as well as Foxp3 protein and antiapoptotic gene expression, and downregulates proapoptotic gene expression. miR-146b knockdown increased the nuclear localization and expression of genes regulated by NF-κB, which was associated with enhanced tTreg survival, proliferation, and suppressive function measured in vitro and in vivo. TRAF6 inhibition had the opposite effects. We conclude that an miR-146b-TRAF6-NF-κB-FoxP3 signaling pathway restrains regulatory T cell survival, proliferation, and suppressor function. In vitro exposure of human tTregs to miR-146b antagomirs can be exploited to improve the clinical efficacy of human adoptive tTreg transfer in a GVHD setting.


Asunto(s)
Antagomirs/genética , Enfermedad Injerto contra Huésped/prevención & control , MicroARNs/genética , FN-kappa B/metabolismo , Linfocitos T Reguladores/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Animales , Apoptosis , Proliferación Celular , Regulación de la Expresión Génica , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , FN-kappa B/genética , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Factor 6 Asociado a Receptor de TNF/genética
20.
Immunity ; 45(1): 83-93, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27438767

RESUMEN

Regulatory T (Treg) cells are important in maintaining self-tolerance and immune homeostasis. The Treg cell transcription factor Foxp3 works in concert with other co-regulatory molecules, including Eos, to determine the transcriptional signature and characteristic suppressive phenotype of Treg cells. Here, we report that the inflammatory cytokine interleukin-6 (IL-6) actively repressed Eos expression through microRNA-17 (miR-17). miR-17 expression increased in Treg cells in the presence of IL-6, and its expression negatively correlated with that of Eos. Treg cell suppressive activity was diminished upon overexpression of miR-17 in vitro and in vivo, which was mitigated upon co-expression of an Eos mutant lacking miR-17 target sites. Also, RNAi of miR-17 resulted in enhanced suppressive activity. Ectopic expression of miR-17 imparted effector-T-cell-like characteristics to Treg cells via the de-repression of genes encoding effector cytokines. Thus, miR-17 provides a potent layer of Treg cell control through targeting Eos and additional Foxp3 co-regulators.


Asunto(s)
Proteínas Portadoras/metabolismo , Colitis/inmunología , Interleucina-6/metabolismo , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Proteínas Portadoras/genética , Células Cultivadas , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Interleucina-6/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Autotolerancia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...