Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Commun Signal ; 15(3): 393-403, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33945104

ABSTRACT

Renal tubular epithelial-myofibroblast transdifferentiation (EMT) plays a central role in the development of renal interstitial fibrosis (RIF). The profibrotic cytokine interleukin (IL)-1 and the IL-1 receptor (IL-1R) also participate in RIF development, and Toll/IL-1R 8 (TIR8), a member of the Toll-like receptor superfamily, has been identified as a negative regulator of IL-1R signaling. However, the functions of TIR8 in IL-1-induced RIF remain unknown. Here, human embryonic kidney epithelial cells (HKC) and unilateral ureteric obstruction (UUO)-induced RIF models on SD rats were used to investigate the functions of TIR8 involving IL-1ß-induced EMT. We showed that IL-1ß primarily triggers TIR8 expression by activating nuclear factor-κB (NF-κB) in HKC cells. Conversely, high levels of TIR8 in HKC cells repress IL-1ß-induced NF-κB activation and inhibit IL-1ß-induced EMT. Moreover, in vitro and in vivo findings revealed that TIR8 downregulation facilitated IL-1ß-induced NF-κB activation and contributed to TGF-ß1-mediated EMT in renal tubular epithelial cells. These results suggested that TIR8 exerts a protective role in IL-1ß-mediated EMT and potentially represents a new target for RIF treatment.

2.
Oncotarget ; 8(33): 54858-54872, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28903387

ABSTRACT

Hyperactivation of mammalian target of rapamycin complex 1 (mTORC1), caused by loss-of-function mutations in either the TSC1 or TSC2 gene, leads to the development of tuberous sclerosis complex (TSC), a benign tumor syndrome with multiple affected organs. mTORC1-mediated inhibition of AKT constrains the tumor progression of TSC, but the exact mechanisms remain unclear. Herein we showed that loss of TSC1 or TSC2 downregulation of platelet-derived growth factor receptor α (PDGFRα) expression was mediated by mTORC1. Moreover, mTORC1 inhibited PDGFRα expression via suppression of forkhead box O3a (FOXO3a)-mediated PDGFRα gene transcription. In addition, ectopic expression of PDGFRα promoted AKT activation and enhanced proliferation and tumorigenic capacity of Tsc1- or Tsc2-null mouse embryonic fibroblasts (MEFs), and vice versa. Most importantly, rapamycin in combination with AG1295, a PDGFR inhibitor, significantly inhibited growth of TSC1/TSC2 complex-deficient cells in vitro and in vivo. Therefore, downregulated FOXO3a/PDGFRα/AKT pathway exerts a protective effect against hyperactivated mTORC1-induced tumorigenesis caused by loss of TSC1/TSC2 complex, and the combination of rapamycin and AG1295 may be a new effective strategy for TSC-associated tumors treatment.

3.
Hum Mol Genet ; 26(2): 407-419, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28013293

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disorder featured with multi-organ benign tumours. Disruption of TSC1/TSC2 complex suppression on mammalian/mechanistic target of rapamycin (mTOR) signalling causes TSC. Hyperactive mTOR-mediated negative feedback regulation of AKT partially contributes to the benign nature of TSC-associated tumours. In this study, we demonstrated that osteopontin (OPN) was dramatically reduced by loss of TSC1/TSC2 complex in Tsc2-null mouse embryonic fibroblasts (MEFs), rat uterine leiomyoma-derived Tsc2-deficient cells, genetically modified mouse TSC models, and clinical samples. TSC1/TSC2 complex upregulation of OPN expression is mediated by transcription factor SOX9 in an mTOR-independent manner. Moreover, ablation of OPN by deficient TSC1/TSC2 complex contributed to inactivation of AKT in TSC cells. Lastly, the abundance of OPN dictated the potency of cell proliferation and tumour development. Therefore, loss of TSC1/TSC2 complex led to mTOR-independent inhibition of AKT at least partially through downregulation of the SOX9-OPN signalling cascade. We suggest that the decreased SOX9-OPN-AKT signalling pathway safeguard against the development of malignant tumours in TSC patients.


Subject(s)
Oncogene Protein v-akt/genetics , Osteopontin/genetics , SOX9 Transcription Factor/genetics , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Proliferation/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Rats , Signal Transduction , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
4.
Oncotarget ; 7(19): 28435-47, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27078846

ABSTRACT

Tuberous sclerosis complex (TSC), caused by loss-of-function mutations in the TSC1 or TSC2 gene, is characterized by benign tumor formation in multiple organs. Hyperactivation of mammalian target of rapamycin complex 1 (mTORC1) is the primary alteration underlying TSC tumors. By analyzing Tsc2-null mouse embryonic fibroblasts (MEFs) and rat uterine leiomyoma-derived Tsc2-null ELT3 cells, we detected evidence for the involvement of cyclooxygenase 2 (COX2) as a downstream target of mTORC1 in the development of TSC tumors. We showed that loss of TSC2 led to decreased COX2 expression through activation of an mTORC1/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Overexpression of COX2 promoted proliferation and tumoral growth of Tsc2-null cells. COX2 knockdown inhibited the proliferation of the control cells. COX2 enhanced Tsc2-null cell growth through upregulation of interleukin-6 (IL-6). In addition, rapamycin in combination with celecoxib, a COX2 inhibitor, strongly inhibited Tsc2-deficient cell growth. We conclude that downregulation of COX2 exerts a protective effect against hyperactivated mTORC1-mediated tumorigenesis caused by the loss of TSC2, and the combination of rapamycin and celecoxib may be an effective new approach to treating TSC.


Subject(s)
Cyclooxygenase 2/biosynthesis , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/genetics , Tuberous Sclerosis/complications , Tumor Suppressor Proteins/deficiency , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinogenesis/metabolism , Celecoxib/pharmacology , Cell Proliferation/drug effects , Down-Regulation , Mice , Neoplasms/metabolism , Rats , Sirolimus/pharmacology , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein
5.
Cancer Lett ; 359(1): 97-106, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25578782

ABSTRACT

Accumulating evidence indicates that mammalian target of rapamycin (mTOR) exerts a crucial role in aerobic glycolysis and tumorigenesis, but the underlying mechanisms remain largely obscure. Results from Tsc1- or Tsc2-null mouse embryonic fibroblasts (MEFs) and human cancer cell lines consistently indicate that the expression of glucose transporter 3 (Glut3) is dramatically up-regulated by mTOR. The rapamycin-sensitive mTOR complex 1 (mTORC1), but not the rapamycin-insensitive mTOR complex 2 (mTORC2), was involved in the regulation of Glut3 expression. Moreover, mTORC1 enhances Glut3 expression through the activation of the IKK/NFκB pathway. Depletion of Glut3 led to the suppression of aerobic glycolysis, the inhibition of cell proliferation and colony formation, and the attenuation of the tumorigenic potential of the cells with aberrantly hyper-activated mTORC1 signaling in nude mice. We conclude that Glut3 is a downstream target of mTORC1, and it is critical for oncogenic mTORC1-mediated aerobic glycolysis and tumorigenesis. Hence Glut3 may be a potential target for therapy against cancers caused by the aberrantly activated mTORC1 signaling.


Subject(s)
Cell Proliferation , Glucose Transporter Type 3/metabolism , Glycolysis , NF-kappa B/metabolism , Neoplasms/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Glucose Transporter Type 3/genetics , Glycolysis/drug effects , HEK293 Cells , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred BALB C , Mice, Nude , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , NF-kappa B/genetics , Neoplasms/genetics , Neoplasms/pathology , Promoter Regions, Genetic , RNA Interference , Rats , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , Time Factors , Transfection , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Burden , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...