Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Autophagy ; 20(2): 460-462, 2024 02.
Article in English | MEDLINE | ID: mdl-37876279

ABSTRACT

Mitophagy, the process of removing damaged mitochondria to promote cell survival, plays a crucial role in cellular functionality. However, excessive, or uncontrolled mitophagy can lead to reduced mitochondrial content that burdens the remaining organelles, triggering mitophagy-mediated cell death. FBXL4 mutations, which affect the substrate-binding adaptor of the CUL1 (cullin 1)-RING ubiquitin ligase complex (CRL1), have been linked to mitochondrial DNA depletion syndrome type 13 (MTDPS13) characterized by reduced mtDNA content and impaired energy production in affected organs. However, the mechanism behind FBXL4 mutation-driven MTDPS13 remain poorly understood. In a recent study, we demonstrate that the CRL1-FBXL4 complex promotes the degradation of BNIP3 and BNIP3L, two key mitophagy cargo receptors. Deficiency of FBXL4 results in a strong accumulation of BNIP3 and BNIP3L proteins and triggers high levels of BNIP3- and BNIP3L-dependent mitophagy. Patient-derived FBXL4 mutations do not affect its interaction with BNIP3 and BNIP3L but impair the assembly of an active CRL1-FBXL4 complex. Furthermore, excessive mitophagy is observed in knockin mice carrying a patient-derived FBXL4 mutation, and in cortical neurons generated from human patient induced pluripotent stem cells (hiPSCs). These findings support the model that the CRL1-FBXL4 complex tightly restricts basal mitophagy, and its dysregulation leads to severe symptoms of MTDPS13.


Subject(s)
Autophagy , Mitochondrial Diseases , Mitophagy , Animals , Humans , Mice , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitophagy/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , F-Box Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism
2.
Redox Biol ; 67: 102872, 2023 11.
Article in English | MEDLINE | ID: mdl-37688978

ABSTRACT

The mechanistic target of the rapamycin (mTOR) pathway, which participates in the regulation of cellular growth and metabolism, is aberrantly regulated in various cancer types. The mTOR complex 2 (mTORC2), which consists of the core components mTOR, Rictor, mSin1, and mLST8, primarily responds to growth signals. However, the coordination between mTORC2 assembly and activity remains poorly understood. Keap1, a major sensor of oxidative stress in cells, functions as a substrate adaptor for Cullin 3-RING E3 ubiquitin ligase (CRL3) to promote proteasomal degradation of NF-E2-related factor 2 (NRF2), which is a transcription factor that protects cells against oxidative and electrophilic stress. In the present study, we demonstrate that Keap1 binds to mLST8 via a conserved ETGE motif. The CRL3Keap1 ubiquitin ligase complex promotes non-degradative ubiquitination of mLST8, thus reducing mTORC2 complex integrity and mTORC2-AKT activation. However, this effect can be prevented by oxidative/electrophilic stresses and growth factor signaling-induced reactive oxygen species (ROS) burst. Cancer-derived Keap1 or mLST8 mutations disrupt the Keap1-mLST8 interaction and allow mLST8 to evade Keap1-mediated ubiquitination, thereby enhancing mTORC2-AKT activation and promoting cell malignancy and remodeling cell metabolism. Our findings provide new insights into the molecular mechanisms of Keap1/mLST8 mutation-driven tumorigenesis by promoting mTORC2-AKT activation, which is independent of the canonical NRF2 pathway.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Mechanistic Target of Rapamycin Complex 2/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , TOR Serine-Threonine Kinases/metabolism , Neoplasms/genetics , Mutation
3.
J Nanobiotechnology ; 21(1): 235, 2023 Jul 22.
Article in English | MEDLINE | ID: mdl-37481565

ABSTRACT

Systemic administration of platinum-based drugs has obvious limitations in the treatment of advanced bladder cancer (BC) owing to lower tumor accumulation and uncontrolled release of chemotherapeutics. There is an urgent need for advanced strategies to overcome the current limitations of platinum-based chemotherapy, to achieve maximal therapeutic outcomes with reduced side effects. In this study, self-polymerized platinum (II)-polydopamine nanocomplexes (PtPDs) were tailored for efficient chemo-photoimmunotherapy of BC. PtPDs with high Pt loading content (11.3%) were degradable under the combination of a reductive tumor microenvironment and near-infrared (NIR) light irradiation, thus controlling the release of Pt ions to achieve efficient chemotherapy. In addition, polydopamine promoted stronger photothermal effects to supplement platinum-based chemotherapy. Consequently, PtPDs provided effective chemo-photothermal therapy of MB49 BC in vitro and in vivo, strengthening the immunogenic cell death (ICD) effect and robust anti-tumoral immunity response. When combined with a PD-1 checkpoint blockade, PtPD-based photochemotherapy evoked systemic immune responses that completely suppressed primary and distant tumor growth without inducing systemic toxicities. Our work provides a highly versatile approach through metal-dopamine self-polymerization for the precise delivery of metal-based chemotherapeutic drugs, and may serve as a promising nanomedicine for efficient and safe platinum-based chemotherapy for BC.


Subject(s)
Nanomedicine , Urinary Bladder Neoplasms , Humans , Polymerization , Indoles , Tumor Microenvironment
4.
Mol Cancer ; 18(1): 170, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31771591

ABSTRACT

BACKGROUND: The gene encoding the E3 ubiquitin ligase substrate-binding adaptor SPOP is frequently mutated in primary prostate cancer, but how SPOP mutations contribute to prostate cancer pathogenesis remains poorly understood. Stress granules (SG) assembly is an evolutionarily conserved strategy for survival of cells under stress, and often upregulated in human cancers. We investigated the role of SPOP mutations in aberrant activation of the SG in prostate cancer and explored the relevanve of the mechanism in therapy resistance. METHODS: We identified SG nucleating protein Caprin1 as a SPOP interactor by using the yeast two hybrid methods. A series of functional analyses in cell lines, patient samples, and xenograft models were performed to investigate the biological significance and clinical relevance of SPOP regulation of SG signaling in prostate cancer. RESULTS: The cytoplasmic form of wild-type (WT) SPOP recognizes and triggers ubiquitin-dependent degradation of Caprin1. Caprin1 abundance is elevated in SPOP-mutant expressing prostate cancer cell lines and patient specimens. SPOP WT suppresses SG assembly, while the prostate cancer-associated mutants enhance SG assembly in a Caprin1-dependent manner. Knockout of SPOP or expression of prostate cancer-associated SPOP mutants conferred resistance to death caused by SG inducers (e.g. docetaxel, sodium arsenite and H2O2) in prostate cancer cells. CONCLUSIONS: SG assembly is aberrantly elevated in SPOP-mutated prostate cancer. SPOP mutations cause resistance to cellular stress induced by chemtherapeutic drug such as docetaxel in prostate cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Docetaxel/pharmacology , Drug Resistance, Neoplasm/genetics , Mutation , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Repressor Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cytoplasmic Granules/metabolism , Fluorescent Antibody Technique , Humans , Male , Models, Biological , Prostatic Neoplasms/drug therapy , Protein Binding , Proteolysis , Stress, Physiological , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Tumour Biol ; 37(7): 9797-806, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26810069

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common primary kidney cancer in adults, and the identification of biomarkers involved in the pathogenesis and prognosis of ccRCC is crucial for early diagnosis and anticancer treatment. In this study, we demonstrate that thioredoxin domain-containing protein 5 (TXNDC5) expression is markedly upregulated in ccRCC tissues in comparison with adjacent non-cancerous tissues through quantitative RT-PCR, Western blotting, and immunohistochemical analyses. Importantly, TXNDC5 expression is negatively correlated with the overall survival of patients. Knockdown of TXNDC5 by siRNAs inhibits the cell growth, migration, and invasion of ccRCC cells as well as sensitizes ccRCC cells to chemotherapeutic drugs, such as Camptothecin and 5-Fluorouracil. Moreover, we used complementary DNA (cDNA) microarray analyses to explore the underlying molecular mechanisms of TXNDC5 in the pathogenesis of ccRCC. We demonstrate that knockdown of TXNDC5 affects the messenger RNA (mRNA) and protein levels of numerous important genes associated with tumorigenesis. In summary, our findings indicate that TXNDC5 performs an essential function in ccRCC pathogenesis and can serve as a novel prognostic marker of ccRCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/secondary , Drug Resistance, Neoplasm , Kidney Neoplasms/pathology , Protein Disulfide-Isomerases/metabolism , Aged , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Blotting, Western , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Case-Control Studies , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Lymphatic Metastasis , Male , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/genetics , RNA, Messenger/genetics , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured
6.
Cancer Lett ; 369(1): 167-74, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26300492

ABSTRACT

Characterization of the exome and genome of carcinoma (ccRCC) by next-generation sequencing identified numerous genetic alternations. BRCA1-associated protein-1 (BAP1) was identified as one of the most frequently mutated genes in ccRCC, suggesting that BAP1 is a potential key driver for ccRCC cancer initiation and progression. However, how BAP1 mutations contribute to ccRCC remains to be elucidated. BAP1 is a nuclear de-ubiquitinating enzyme and cleaves the ubiquitin chain from the substrates. Here, we identified MCRS1 as a bona fide substrate for BAP1. MCRS1 is a component of the centrosome proteins, and plays an essential role in spindle assembly. BAP1 binds to MCRS1 and stabilizes MCRS1 by de-ubiquitination. BAP1 contributes to chromosome stability partially via MCRS1. A positive correlation was identified between BAP1 and MCRS1 expression in ccRCC tissues. Both BAP1 loss and MCRS1 down-regulation in ccRCC were associated with adverse clinicopathological features. This study revealed a novel mechanism for BAP1 involved in MCRS1 stability regulation, and provided insight in understanding the relationship between BAP1 mutations and chromosome instability in ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Chromosomal Instability , Kidney Neoplasms/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/physiology , Ubiquitin Thiolesterase/physiology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Down-Regulation , Female , HEK293 Cells , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Middle Aged , Protein Interaction Domains and Motifs , Protein Stability , Ubiquitination
7.
Biochem Biophys Res Commun ; 464(1): 147-53, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26102039

ABSTRACT

Ubiquitin-like with PHD and RING finger domain 1 (UHRF1) is a multi-domain ubiquitin E3 ligase that plays critical roles in regulation of DNA methylation and histone ubiquitination. In this study, we found UHRF1 is frequently overexpressed in human clear cell Renal Cell Carcinoma (ccRCC) tissues both at mRNA and protein levels. We showed that UHRF1 directly interacts with p53 both in vivo and in vitro. A new domain (PD) in UHRF1 was required for interaction with p53. We found that UHRF1 down-regulates p53 transactivation activity which was depends on the ubiquitin E3 ligase function. UHRF1 can promote non-degradative ubiquitination of p53, suppress p53 pathway activation and p53-dependent apoptosis in ccRCC cells. Together, our study suggests that UHRF1, which overexpressed ccRCC, may act as a p53 regulator, suppress p53 pathway activation and help ccRCC cells to escape from p53-dependent apoptosis.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , DNA Damage , HEK293 Cells , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases , Ubiquitination
8.
World J Surg Oncol ; 13: 175, 2015 May 08.
Article in English | MEDLINE | ID: mdl-25951903

ABSTRACT

BACKGROUND: Renal cell carcinoma (RCC) is characterized by inherent resistance to chemotherapy. Earlier studies demonstrated that microRNAs (miRNAs) might be involved in the chemosensitivity of cancers. MicroRNA let-7, a putative tumor suppressor, is dysregulated in many cancers. Our study aims to investigate the exact role of let-7 in chemotherapy sensitivity of 5-fluorouracil (5-FU) in RCC. METHODS: The clinical significance of let-7b and let-7c expression in surgically resected specimens was assessed by qRT-PCR. Cell proliferation assay and colony formation assay were used to assess the survival of 786-O cells treated with let-7b or let-7c combined with 5-FU. Western blot was used to detect the expression of Akt2 and caspase-7. Luciferase assay was used to detect the direct binding of let-7b and let-7c to the 3'-untranslated region (UTR) of Akt2. RESULTS: Expression of let-7b and let-7c was significantly decreased in 32 paired clear cell renal cell carcinoma tissue specimens and the dysregulation of let-7b was associated with pathological grade. Transfection of let-7b or let-7c combined with 5-FU inhibited proliferation and potentiated the antitumor efficacies of 5-FU at tolerated concentration. let-7b and let-7c suppressed the luciferase activity of reporter plasmid containing the 3'-UTR of Akt2. Overexpression of let-7b and let-7c reduced Akt2 expression, and Akt2 inhibition enhanced the sensitivity to 5-FU by affecting apoptotic pathway. CONCLUSIONS: Expression of let-7b and let-7c was frequently decreased in clear cell renal cell carcinoma tissues. The dysregulation of let-7b and let-7c may be involved in chemoresistance of RCC cells to 5-FU by down-regulating Akt2.


Subject(s)
Carcinoma, Renal Cell/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Kidney Neoplasms/drug therapy , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/metabolism , 3' Untranslated Regions/genetics , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Cell Proliferation/drug effects , Female , Fluorouracil/pharmacology , Follow-Up Studies , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
Oncol Lett ; 7(5): 1689-1694, 2014 May.
Article in English | MEDLINE | ID: mdl-24765202

ABSTRACT

MicroRNA-34a (miR-34a) is a tumor suppressive microRNA, which induces G1 arrest, apoptosis and senescence by repressing the expression of multiple oncogenes. This study aimed to investigate the biological function and molecular mechanisms of miR-34a in human renal cell carcinoma (RCC) cells. Quantitative polymerase chain reaction (qPCR) revealed that miR-34a expression was significantly downregulated in eight of the 10 (80%) RCC tissues compared with adjacent normal tissues. In RCC cell lines, several other target genes of miR-34a were dysregulated at the mRNA level when the expression of miR-34a was elevated. In addition, western blot analysis and qPCR revealed that forced expression of miR-34a downregulated the expression of Notch1 at the protein and mRNA level. The Cell Counting Kit-8 identified that transient forced expression of miR-34a inhibited cell growth and resulted in cell cycle arrest, which was evaluated by flow cytometry. Our data demonstrated that miR-34a inhibits cell proliferation by downregulating Notch1 in RCC cell lines.

SELECTION OF CITATIONS
SEARCH DETAIL
...