Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Cell Death Dis ; 12(1): 134, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33510128

ABSTRACT

Cancer cachexia is a metabolic disorder characterized by skeletal muscle wasting and white adipose tissue browning. Specific functions of several hormones, growth factors, and cytokines derived from tumors can trigger cachexia. Moreover, adipose tissue lipolysis might explain weight loss that occurs owing to cachexia. Extracellular vesicles (EVs) are involved in intercellular communication. However, whether EVs participate in lipolysis induced by cancer cachexia has not been thoroughly investigated. Using Lewis lung carcinoma (LLC) cell culture, we tested whether LLC cell-derived EVs can induce lipolysis in 3T3-L1 adipocytes. EVs derived from LLC cells were isolated and characterized biochemically and biophysically. Western blotting and glycerol assay were used to study lipolysis. LLC cell-derived EVs induced lipolysis in vivo and vitro. EVs fused directly with target 3T3-L1 adipocytes and transferred parathyroid hormone-related protein (PTHrP), activating the PKA signaling pathway in 3T3-L1 adipocytes. Blocking PTHrP activity in LLC-EVs using a neutralizing antibody and by knocking down PTHR expression prevented lipolysis in adipocytes. Inhibiting the PKA signaling pathway also prevents the lipolytic effects of EVs. In vivo, suppression of LLC-EVs release by knocking down Rab27A alleviated white adipose tissue browning and lipolysis. Our data showed that LLC cell-derived EVs induced adipocyte lipolysis via the extracellular PTHrP-mediated PKA pathway. Our data demonstrate that LLC-EVs induce lipolysis in vitro and vivo by delivering PTHrP, which interacts with PTHR. The lipolytic effect of LLC-EVs was abrogated by PTHR knockdown and treatment with a neutralizing anti-PTHrP antibody. Together, these data show that LLC-EV-induced lipolysis is mediated by extracellular PTHrP. These findings suggest a novel mechanism of lipid droplet loss and identify a potential therapeutic strategy for cancer cachexia.


Subject(s)
Adipose Tissue, Brown/physiology , Cachexia/physiopathology , Extracellular Vesicles/pathology , Lipolysis/physiology , Parathyroid Hormone-Related Protein/metabolism , Animals , Carcinoma, Lewis Lung , Cell Differentiation , Humans , Male , Mice
3.
J Cancer ; 11(5): 1151-1161, 2020.
Article in English | MEDLINE | ID: mdl-31956361

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is one of the most common malignant tumors of the urinary system and has a poor response to radiotherapy and chemotherapy. To date, it is urgent to find effective biomarkers for the prevention and treatment of ccRCC. The occurrence and development of ccRCC is closely related to metabolic disturbances. Palmitoyl protein thioesterase 2 (PPT2) is a lysosomal thioesterase which is highly associated with metabolism, and it has never been studied in ccRCC. In this study, we first revealed PPT2 is significantly downregulated in ccRCC, and its expression level is highly correlated with clinicopathological parameters of ccRCC patients. Our ROC curve analyses evaluated the potential of PPT2 as a novel diagnostic marker and prognostic factor. Functional experiment results showed overexpression of PPT2 represses the proliferation, migration and invasion of ccRCC cells in vitro. Mechanistic investigations demonstrated that overexpression of PPT2 represses the ccRCC progression by reducing epithelial-to-mesenchymal transition (EMT). In conclusion, PPT2 is downregulated in ccRCC. Decreased PPT2 expression may be considered as a novel diagnostic marker and prognostic factor and serve as a therapeutic target for ccRCC.

4.
J Pineal Res ; 67(4): e12607, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31469445

ABSTRACT

Metabolic adaptations are emerging hallmarks of cancer progression and cellular transformation. Clear cell renal cell carcinoma (ccRCC) is a metabolic disease defined histologically by lipid accumulation and lipid storage, which promote tumor cell survival; however, the significance of eliminating the lipid remains unclear. Here, we demonstrate that melatonin activates transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1A (PGC1A) and uncoupling protein 1 (UCP1)-dependent lipid autophagy and a lipid browning program to elicit a catabolic state called "tumor slimming," thus suppressing tumor progression. Metabolic coregulator data analysis revealed that PGC1A expression was decreased in ccRCC tissues versus normal tissues, and poor patient outcome was associated with lower expression of PGC1A in The Cancer Genome Atlas (TCGA-KIRC). PGC1A was downregulated in ccRCC and associated with disease progression. Restoration of PGC1A expression by melatonin in ccRCC cells significantly repressed tumor progression and eliminated the abnormal lipid deposits. Furthermore, a phenomenon called "tumor slimming" was observed, in which tumor cell volume was reduced and lipid droplets transformed into tiny pieces. Additional studies indicated that melatonin promoted "tumor slimming" and suppressed ccRCC progression through PGC1A/UCP1-mediated autophagy and lipid browning. During this process, autophagy and lipid browning eliminate lipid deposits without providing energy. These studies demonstrate that the novel "tumor slimming" pathway mediated by melatonin/PGC1A/UCP1 exhibits prognostic potential in ccRCC, thus revealing the significance of monitoring and manipulating this pathway for cancer therapy.


Subject(s)
Autophagic Cell Death/drug effects , Lipid Metabolism/drug effects , Melatonin/pharmacology , Neoplasm Proteins/metabolism , Neoplasms , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Uncoupling Protein 1/metabolism , Animals , Cell Line, Tumor , Humans , Lipid Droplets/metabolism , Lipid Droplets/pathology , Mice , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Uncoupling Protein 1/genetics , Xenograft Model Antitumor Assays
5.
Adv Sci (Weinh) ; 6(10): 1801862, 2019 May 17.
Article in English | MEDLINE | ID: mdl-31131187

ABSTRACT

Emerging evidence has highlighted the important role of abnormal lipid accumulation in cancer development and progression, but the mechanism for this phenomenon remains unclear. Here, it is demonstrated that phospholipase C-like 1/uncoupling protein 1 (PLCL1)/(UCP1)-mediated lipid browning promotes tumor cell "slimming" and represses tumor progression. By screening three independent lipid metabolism-related gene sets in clear cell renal cell carcinoma (ccRCC) and analyzing the TCGA database, it is found that PLCL1 predicted a poor prognosis and was downregulated in ccRCC. Restoration of PLCL1 expression in ccRCC cells significantly represses tumor progression and reduces abnormal lipid accumulation. Additionally, a phenomenon called tumor cell "slimming," in which tumor cell volume is reduced and lipid droplets are transformed into tiny pieces, is observed. Further studies show that PLCL1 promotes tumor cell "slimming" and represses tumor progression through UCP1-mediated lipid browning, which consumes lipids without producing ATP energy. Mechanistic investigations demonstrate that PLCL1 improves the protein stability of UCP1 by influencing the level of protein ubiquitination. Collectively, the data indicate that lipid browning mediated by PLCL1/UCP1 promotes tumor cell "slimming" and consumes abnormal lipid accumulation, which represses the progression of ccRCC. Tumor cell "slimming" offers a promising new concept and treatment modality against tumor development and progression.

6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(8): 1091-1102, 2019 08.
Article in English | MEDLINE | ID: mdl-31002945

ABSTRACT

Cancer-associated cachexia (CAC) constitutes a metabolic dysfunction characterized by systemic inflammation and body weight loss. Muscle atrophy and adipose tissue lipolysis might explain weight loss in CAC. Specific functions of numerous hormones and cytokines derived from tumours can provoke cachexia. Extracellular vesicles (EVs) can be involved in intercellular communication. However, whether EVs participate in this process has not been investigated thoroughly. Using Lewis lung carcinoma (LLC) cell cultures, we tested whether LLC-derived EVs induced C2C12 myotube atrophy and 3T3-L1 adipocyte lipolysis. EVs derived from LLC cells and serum from patients with lung cancer, non-lung cancer controls, tumour-bearing mice, and non-tumour-bearing control mice were isolated and characterized biochemically and biophysically. LLC cell-derived EVs induced dose-dependent effects of atrophy in C2C12 myotubes and lipolysis in 3T3-L1 adipocytes. Mechanistically, EVs directly fused with target C2C12 myotubes and 3T3-L1 adipocytes, and transferred interleukin-6 (IL-6) activates the STAT3 signalling pathway in C2C12 myotubes and 3T3-L1 adipocytes. Neutralization of extracellular IL-6 prevented the atrophy and lipolysis effects of EVs. Inhibiting the STAT3 signalling pathway also prevented the atrophy and lipolysis effects of EVs. PKH67-labelled (PKH 67 is a lipid dye that can be used to label extracellular vesicles) LLC-EVs were readily internalized into myotubes and adipocytes. Our data showed that LLC cell-derived EVs induced myotube atrophy and adipocyte lipolysis via the extracellular IL-6-mediated STAT3 pathway in target cells. These findings represent a potentially novel basis for further research in this field towards identifying targets and developing strategies for maintaining weight in CAC.


Subject(s)
Adipocytes/metabolism , Extracellular Vesicles/physiology , Interleukin-6/pharmacology , Lung Neoplasms/pathology , Muscle Fibers, Skeletal/pathology , STAT3 Transcription Factor/metabolism , 3T3-L1 Cells , Animals , Atrophy/chemically induced , Cachexia/etiology , Carcinoma, Lewis Lung/pathology , Extracellular Vesicles/metabolism , Humans , Lipolysis/drug effects , Mice , Muscle Fibers, Skeletal/metabolism , Tumor Cells, Cultured
7.
Biochem Biophys Res Commun ; 506(1): 122-129, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30340833

ABSTRACT

Cancer-associated cachexia (CAC) is a disorder characterized by unintended weight loss due to skeletal muscle wasting and adipose tissue loss. Although muscle atrophy in this condition has been well studied, the mechanisms underlying adipose tissue loss, which include browning, have not been investigated in detail. In this respect, though recent studies have shown that exosomes from cancer cells can promote lipolysis, the link between exosomes from cancer cells and CAC has not been clearly established. In this study, we investigate if exosomes from Lewis lung carcinoma (LLC) cells can induce lipolysis in vitro (in 3T3-L1 adipocytes) and in vivo (in LLC tumor-bearing mice). We find that exosomes from LLC cells do induce lipolysis in 3T3-L1 adipocytes and that the white adipose tissues of mice with LLC tumors show clear signs of lipolysis. We also find that this lipolysis can be inhibited using the neutral sphingomyelinase inhibitor GW4869. Our results indicate that GW4869 not only inhibits exosome generation and release from LLC cells, but can also inhibit lipolysis induced by LLC-derived exosomes in 3T3-L1 adipocytes. Furthermore, we also demonstrate that LLC tumor-bearing mice treated with GW4869 did not develop CAC. In summary, our results suggest that inhibiting exosome generation and release can inhibit lipolysis and adipose tissue browning, and may be useful as a novel strategy for treating CAC.


Subject(s)
Adipocytes/drug effects , Aniline Compounds/pharmacology , Benzylidene Compounds/pharmacology , Cachexia/prevention & control , Carcinoma, Lewis Lung/metabolism , Enzyme Inhibitors/pharmacology , Exosomes/drug effects , Lipolysis/drug effects , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Biological Transport , Cachexia/complications , Cachexia/metabolism , Cachexia/pathology , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/pathology , Cell Differentiation , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Exosomes/metabolism , Exosomes/pathology , Male , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured
8.
Cell Physiol Biochem ; 48(3): 1075-1087, 2018.
Article in English | MEDLINE | ID: mdl-30041179

ABSTRACT

BACKGROUND/AIMS: MIAT is a long noncoding RNA (lncRNA) involved in cell proliferation and the development of tumor. However, the exact effects and molecular mechanisms of MIAT in clear cell renal cell carcinoma (ccRCC) progression are still unknown. METHODS: We screened the lncRNAs' profile of ccRCC in The Cancer Genome Atlas database, and then examined the expression levels of lncRNA MIAT in 45 paired ccRCC tissue specimens and in cell lines by q-RT-PCR. MTS, colony formation, EdU, and Transwell assays were performed to examine the effect of MIAT on proliferation and metastasis of ccRCC. Western blot and luciferase assays were performed to determine whether MIAT can regulate Loxl2 expression by competitively binding miR-29c in ccRCC. RESULTS: MIAT was up-regulated in ccRCC tissues and cell lines. High MIAT expression correlated with worse clinicopathological features and shorter survival rate. Functional assays showed that knockdown of MIAT inhibited renal cancer cell proliferation and metastasis in vitro and in vivo. Luciferase and western blot assays further confirmed that miR-29c binds with MIAT. Additionally, the correlation of miR-29c with MIAT and Loxl2 was further verified in patients' samples. CONCLUSION: Our data indicated that MIAT might be an oncogenic lncRNA that promoted proliferation and metastasis of ccRCC, and could be a potential therapeutic target in human ccRCC.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Amino Acid Oxidoreductases/chemistry , Amino Acid Oxidoreductases/genetics , Animals , Antagomirs/metabolism , Binding, Competitive , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Proliferation , Female , Humans , Kidney Neoplasms/genetics , Male , Mice , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Metastasis , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Up-Regulation
10.
Cell Physiol Biochem ; 43(6): 2420-2433, 2017.
Article in English | MEDLINE | ID: mdl-29073615

ABSTRACT

BACKGROUND/AIMS: We previously performed microRNA (miRNA) microarray to identify effective indicators of clear cell renal cell carcinoma (ccRCC) tissue samples and preoperative/postoperative plasma in which we identified miR-144-3p as an oncomiRNA. However, the molecular mechanism of miR-144-3p remains unclear. This study aims to explore the roles of miR-144-3p in the invasion, migration and Sunitinib-resistance in ccRCC and to elucidate the underlying mechanisms. METHODS: Gain and loss of function approaches were used to investigate the cell proliferation, cycle distribution, clonogenicity, migration, invasion, chemosensitivity of miR-144-3p in vitro. The xenograft model was used to assess the effects of miR-144-3p overexpression on tumorigenesis. Bioinformatics analysis and dual-luciferase reporter assay were used to indentify AT-rich interactive domain 1A (ARID1A) as a direct target gene of miR-144-3p. Quantitative RT-PCR, Western blotting, and immunohistochemical (IHC) staining were used to explore ARID1A expression level of the mRNA and protein. RESULTS: We found that miR-144-3p overexpression enhanced cell proliferation, clonogenicity, migration, invasion, and chemoresistance in ccRCC cells. Notably, the oncotumor activities of miR-144-3p were mediated by repressing the expression of ARID1A. The downregulation of ARIDIA could promote the function of miR-144-3p in cell proliferation, metastasis and chemoresistance. Consistently, ARID1A mRNA and protein levels were decreased in ccRCC and in nude mice, and they negatively correlated with miR-144-3p. CONCLUSION: Higher miR-144-3p may enhance malignancy and resistance to Sunitinib in ccRCC by targeting ARID1A, the observations may uncover novel strategies of ccRCC treatment.


Subject(s)
Carcinoma, Renal Cell/pathology , Drug Resistance, Neoplasm , Kidney Neoplasms/pathology , MicroRNAs/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Base Sequence , Carcinogenesis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins , Down-Regulation , Humans , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Male , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mutagenesis , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Pyrroles/therapeutic use , RNA Interference , RNA, Small Interfering/metabolism , Sequence Alignment , Sunitinib , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transplantation, Heterologous , Up-Regulation
11.
Exp Cell Res ; 358(2): 269-278, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28684115

ABSTRACT

Caveolin-1 (CAV1) has been identified to be up-regulated in many cancers, including clear cell renal cell carcinoma (ccRCC). However, its potential function is still unclear in ccRCC. In this study, we demonstrated that CAV1 was frequently overexpressed in renal cell carcinoma tissues and cells, and was significantly associated with various clinicopathological parameters. In addition, high CAV1 expression was associated with poor disease-free survival (DFS) rate and could serve as a useful diagnostic indicator in ccRCC patients with different clinicopathological stages. Functional experiments demonstrated that CAV1 knockdown inhibited cell migration and invasion, whereas overexpression of CAV1 promoted cell migration and invasion in ccRCC. Moreover, CAV1 expression was up-regulated in sunitinib-resistant renal cancer cell lines, and its overexpression promoted sunitinib resistance. In general, our results confirm that CAV1 plays an important role in the metastasis of kidney cancer and induces sunitinib resistance, so CAV1 function suppression may become a promising clinical treatment strategy during renal cell carcinoma metastasis and sunitinib resistance.


Subject(s)
Carcinoma, Renal Cell/metabolism , Caveolin 1/metabolism , Cell Movement/physiology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/genetics , Indoles/pharmacology , Kidney Neoplasms/metabolism , Pyrroles/pharmacology , Adult , Aged , Aged, 80 and over , Caveolin 1/genetics , Cell Proliferation/genetics , Cell Proliferation/physiology , Female , Gene Knockdown Techniques/methods , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Sunitinib , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...