Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cancer Cell Int ; 22(1): 101, 2022 Mar 03.
Article in English | MEDLINE | ID: mdl-35241075

ABSTRACT

BACKGROUND: Emerging evidence suggests that LMNB1 is involved in the development of multiple cancer types. However, there is no study reporting the potential role of LMNB1 in a systematic pan-cancer manner. METHODS: The gene expression level and potential oncogenic roles of LMNB1 in The Cancer Genome Atlas (TCGA) database were analyzed with Tumor Immune Estimation Resource version 2 (TIMER2.0), Gene Expression Profiling Interactive Analysis version 2 (GEPIA2), UALCAN and Sangerbox tools. Pathway enrichment analysis was carried out to explore the possible mechanism of LMNB1 on tumorigenesis and tumor progression. The therapeutic effects of LMNB1 knockdown combined with PARP inhibition on human cancers were further investigated in vitro. RESULTS: LMNB1 upregulation is generally observed in the tumor tissues of most TCGA cancer types, and is verified in kidney renal clear cell carcinoma using clinical specimens of our institute. High level of LMNB1 expression usually predicts poor overall survival and disease free survival for patients with tumors. Mechanically, LMNB1 level is positively correlated with CD4+ Th2 cell infiltration and DNA homologous recombination repair gene expression. In vitro experiments reveal that targeting LMNB1 has a synergistic effect on prostate cancer with PARP inhibitor treatment. CONCLUSIONS: LMNB1 is a biomarker of CD4+ Th2 cell infiltration and DNA homologous recombination repair in human cancers. Blockage of LMNB1 combined with PARP inhibitor treatment could be a promising therapeutic strategy for patients with cancers.

2.
J Nanobiotechnology ; 18(1): 124, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32887622

ABSTRACT

BACKGROUND: Chemotherapy is a standard cancer treatment which uses anti-cancer drugs to destroy or slow the growth of cancer cells. However, chemotherapy has limited therapeutic effects in bladder cancer. One of the reasons of this resistance to chemotherapy is that higher levels of glutathione in invasive bladder cancer cells. We have fabricated nanoparticles that respond to high concentrations of glutathione and near-infrared laser irradiation in order to increase the drug accumulation at the tumor sites and combine chemotherapy with photothermal therapy to overcome the challenges of bladder cancer treatment. METHODS: The DOX&IR780@PEG-PCL-SS NPs were prepared by co-precipitation method. We investigated the tumor targeting capability of NPs in vitro and in vivo. The orthotopic bladder cancer model in C57BL/6 mice was established for in vivo study and the photothermal effects and therapeutic efficacy of NPs were evaluated. RESULTS: The DOX&IR780@PEG-PCL-SS NPs were synthesized using internal cross-linking strategy to increase the stability of nanoparticles. Nanoparticles can be ingested by tumor cells in a short time. The DOX&IR780@PEG-PCL-SS NPs have dual sensitivity to high levels of glutathione in bladder cancer cells and near-infrared laser irradiation. Glutathione triggers chemical structural changes of nanoparticles and preliminarily releases drugs, Near-infrared laser irradiation can promote the complete release of the drugs from the nanoparticles and induce a photothermal effect, leading to destroying the tumor cells. Given the excellent tumor-targeting ability and negligible toxicity to normal tissue, DOX&IR780@PEG-PCL-SS NPs can greatly increase the concentration of the anti-cancer drugs in tumor cells. The mice treated with DOX&IR780@PEG-PCL-SS NPs have a significant reduction in tumor volume. The DOX&IR780@PEG-PCL-SS NPs can be tracked by in vivo imaging system and have good tumor targeting ability, to facilitate our assessment during the experiment. CONCLUSION: A nanoparticle delivery system with dual sensitivity to glutathione and near-infrared laser irradiation was developed for delivering IR780 and DOX. Chemo-photothermal synergistic therapy of both primary bladder cancer and their metastases was achieved using this advanced delivery system.


Subject(s)
Antineoplastic Agents/pharmacology , Muscle Neoplasms/drug therapy , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polymers/chemistry , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Drug Delivery Systems , Drug Therapy/methods , Humans , Infrared Rays , Laser Therapy , Lasers , Mice , Mice, Inbred C57BL , Muscle Neoplasms/pathology , Muscle Neoplasms/radiotherapy , Muscles/drug effects , Phototherapy/methods , Polyethylene Glycols , Sensitivity and Specificity , Succinimides , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/radiotherapy
3.
J Cell Biochem ; 120(9): 15924-15932, 2019 09.
Article in English | MEDLINE | ID: mdl-31069875

ABSTRACT

Recent studies have suggested that miR-30e-5p is dysregulated in several human carcinomas; however, the mechanism of miR-30e-5p in bladder cancer (BCa) remains unknown. Here, we confirmed that the expression of miR-30e-5p was decreased in human BCa specimens and cell lines by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). Upregulation of miR-30e-5p decreased the proliferation and migration in T24 and UM-UC-3 cells. Metadherin (MTDH) was a potential target for miR-30e-5p through bioinformatics analysis. Dual-luciferase assays were conducted to validate the interaction between miR-30e-5p and MTDH, which demonstrates that the relative luciferase activity was significantly downregulated after transfected miR-30e-5p mimic compared with control mimic in 293T cells. We also detected that whether silencing of MTDH by using small interfering(si)-MTDH matched effects caused by miR-30e-5p overexpression in BCa cells lines by Cell Counting Kit-8 (CCK-8), colony formation, and transwell assay, and we found the effects of silencing of MTDH same as miR-30e-5p overexpression. Furthermore, we verified that the restoration of MTDH in miR-30e-5p-overexpressed BCa cells rescued the inhibitory effects of miR-30e-5p. In conclusion, these results demonstrated that miR-30e-5p may inhibit BCa cells growth and invasiveness by targeting MTDH and may be a promising therapeutic agent for treating clinical BCa patients.


Subject(s)
Down-Regulation , Membrane Proteins/genetics , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Urinary Bladder Neoplasms/genetics , 3' Untranslated Regions , Aged , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged
4.
Oncol Lett ; 25(6): 274, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37216166

ABSTRACT

Most patients with renal cancer will develop resistance to sorafenib therapy and will therefore exhibit disease progression. Effective therapies for these patients are extremely limited. Cyclooxygenase-2 (COX-2) promotes the malignant transformation of cancer cells and drug resistance. The potential of COX-2 inhibitor (celecoxib) administration in combination with sorafenib for the treatment of renal cancer is unclear. The present study demonstrated that sorafenib rapidly increased the expression of COX-2 in renal cancer cells, as determined using reverse transcription-quantitative PCR and western blotting. The results of the MTT assay and cell apoptosis experiment demonstrated that the cytotoxicity of sorafenib was also affected by COX-2 expression and celecoxib enhanced the cytotoxicity of sorafenib against renal cell carcinoma. Immunofluorescence analysis indicated that sorafenib induced the formation of stress granules (SGs) in renal cancer cells. In addition, COX-2 expression was associated with the formation of SGs, and SGs could capture and stabilize COX-2 mRNAs in renal cancer cells; this was confirmed using RNA fluorescence in situ hybridization and an actinomycin D chase experiment. The protective effect of SGs was further demonstrated in cell experiments and xenograft tumor models. Thus, the present study indicated that the use of celecoxib may significantly enhance the sensitivity of renal cancer cells to sorafenib and improve efficacy. Sorafenib-induced SGs may contribute to critical events that promote COX-2 expression and survival in renal cancer cells. Therefore, the present study may provide novel ideas for the treatment of renal cancer.

5.
J Control Release ; 363: 221-234, 2023 11.
Article in English | MEDLINE | ID: mdl-37717657

ABSTRACT

Checkpoint blockade immunotherapy (CBI) have exhibited remarkable benefits for cancer therapy. However, the low responsivity of CBI hinders its application in treatment of bladder cancer. Ferroptosis shows potential for increasing the responsivity of CBI by inducing immunogenic cell death (ICD) process. Herein, we developed a mitochondrial-targeted liposome loaded with brequinar (BQR) (BQR@MLipo) for enhancing the mitochondrial-related ferroptosis in bladder cancer in situ. It could be found that BQR@MLipo could selectively accumulate into mitochondria and inactivate dihydroorotate dehydrogenase (DHODH), which induced extensive mitochondrial lipid peroxidation and ROS, finally triggering ferroptosis of bladder cancer cells to boost the release of intracellular damage-associated molecular patterns (DAMPs) such as calreticulin (CRT), adenosine triphosphate (ATP), high mobility group box 1 (HMGB1). In addition, BQR@MLipo further promoted the release of mtDNA into the cytoplasm to activate the cGAS-STING pathway for the secretion of IFN-ß, which would increase the cross-presentation of antigens by dendritic cells and macrophage phagocytosis. Furthermore, the in vivo studies revealed that BQR@MLipo could remarkably accumulate into the bladder tumor and successfully initiate the infiltration of CD8+ T cells into tumor microenvironment for enabling efficient CBI to inhibit bladder tumor growth. Therefore, BQR@MLipo may represent a clinically promising modality for enhancing CBI in bladder tumor.


Subject(s)
Ferroptosis , Urinary Bladder Neoplasms , Humans , CD8-Positive T-Lymphocytes , Immune Checkpoint Inhibitors , Liposomes , Immunotherapy , Urinary Bladder Neoplasms/drug therapy , Mitochondria , Cell Line, Tumor , Tumor Microenvironment
6.
Oncogenesis ; 12(1): 47, 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37783676

ABSTRACT

Lymph node (LN) metastasis is one of the key prognostic factors in bladder cancer, but its underlying mechanisms remain unclear. Here, we found that elevated expression of WD repeat domain 4 (WDR4) in bladder cancer correlated with worse prognosis. WDR4 can promote the LN metastasis and proliferation of bladder cancer cells. Mechanistic studies showed that WDR4 can promote the nuclear localization of DEAD-box helicase 20 (DDX20) and act as an adaptor to bind DDX20 and Early growth response 1 (Egr1), thereby inhibiting Egr1-promoted transcriptional expression of arrestin beta 2 (ARRB2) and ultimately contributing to the progression of bladder cancer. Immunohistochemical analysis confirmed that WDR4 expression is also an independent predictor of LN metastasis in bladder cancer. Our results reveal a novel mechanism of LN metastasis and progression in bladder cancer and identify WDR4 as a potential therapeutic target for metastatic bladder cancer.

7.
Cancer Lett ; 578: 216463, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37866544

ABSTRACT

The role of long non-coding RNA (lncRNA) in the progression of renal cell carcinoma (RCC) remains further study. Whether lncRNA may be used to predict the immunotherapy efficacy of RCC is less studied. In this study, LINC00926 was found to be mainly located in cytoplasm by FISH and RNA nuclear-cytoplasmic fractionation. Downregulation of LINC00926 in RCC cell lines inhibited the progression and metastasis of RCC cells. RNA pull-down assay and dual-luciferase reporter assay demonstrated that LINC00926 functioned as miR-30a-5p sponge to facilitate SOX4 expression. LINC00926 overexpression in BALB/c mice enhanced PD-L1 surface expression and resulted in immune escape. Mechanistic investigations showed that LINC00926 competitively bound to Lyn, leading to the inhibition of CBL-mediated ubiquitination and degradation, and stabilized Lyn, contributing to the activation of IFNγ-JAK2-STAT1 signaling pathway. Moreover, LINC00926, together with PD-L1 or PD-1 expression, may predict the overall survival in RCC patients. Therefore, LINC00926 has the potential to be a novel therapeutic target and a biomarker to predict ICB immunotherapy response in RCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , MicroRNAs , RNA, Long Noncoding , Animals , Humans , Mice , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Janus Kinase 2/metabolism , Kidney Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , SOXC Transcription Factors/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
8.
Redox Biol ; 68: 102956, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37977044

ABSTRACT

The specific mechanism of clear cell renal cell carcinoma (ccRCC) progression, a pathological type that accounts for the highest proportion of RCC, remains unclear. In this study, bioinformatics analysis of scRNA-seq dataset in ccRCC revealed that MIOX was a gene specifically down-regulated in tumor epithelial cells of ccRCC. Analysis of the TCGA database further validated the association between decreased MIOX mRNA levels and ccRCC malignant phenotype and poor prognosis. Immunohistochemistry indicated the down-regulation of MIOX in ccRCC tissues compared to paired adjacent renal tissues, with further down-regulation of MIOX in the primary tumors of patients with primary metastasis compared to those without metastasis. Also, patients with low expression of MIOX showed shorter metastasis-free survival (MFS) compared to those with high MIOX expression. In vitro results showed that overexpression of MIOX in ccRCC cells inhibited the proliferation, migration and invasion and promoted apoptosis. Mechanistically, up-regulation of MIOX inhibited autophagy to elevate the levels of ROS, and thus suppressed STAT3/c-Myc-mediated epithelial-mesenchymal transition in ccRCC cells. In vivo data further confirmed that increased MIOX expression suppressed the growth and proliferation of RCC cells and reduced the ability of RCC cells to form metastases in the lung. This study demonstrates that MIOX is an important regulatory molecule of ccRCC, which is conducive to understanding the potential molecular mechanism of ccRCC progression.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Autophagy/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
9.
Adv Healthc Mater ; 12(21): e2300191, 2023 08.
Article in English | MEDLINE | ID: mdl-37031357

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs), two immunosuppressive myeloid components within the tumor microenvironment (TME), represent fundamental barriers in cancer immunotherapy, whereas current nanomedicines rarely exert dual modulatory roles on these cell types simultaneously. Reactive oxygen species (ROS) not only mediates MDSC-induced immunosuppression but also triggers differentiation and polarization of M2-TAMs. Herein, an ROS scavenging nanozyme, Zr-CeO, with enhanced superoxide dismutase- and catalase-like activities for renal tumor growth inhibition is reported. Mechanistically, intracellular ROS scavenging by Zr-CeO significantly attenuates MDSC immunosuppression via dampening the unfolded protein response, hinders M2-TAM polarization through the ERK and STAT3 pathways, but barely affects neoplastic cells and cancer-associated fibroblasts. Furthermore, Zr-CeO enhances the antitumor effect of PD-1 inhibition in murine renal and breast tumor models, accompanied with substantially decreased MDSC recruitment and reprogrammed phenotype of TAMs in the tumor mass. Upon cell isolation, reversed immunosuppressive phenotypes of MDSCs and TAMs are identified. In addition, Zr-CeO alone or combination therapy enhances T lymphocyte infiltration and IFN-γ production within the TME. Collectively, a promising strategy to impair the quantity and function of immunosuppressive myeloid cells and sensitize immunotherapy in both renal and breast cancers is provided.


Subject(s)
Immunosuppression Therapy , Neoplasms , Animals , Mice , Reactive Oxygen Species/metabolism , Immune Tolerance , Myeloid Cells/metabolism , Neoplasms/metabolism , Tumor Microenvironment , Immunotherapy
10.
Oncoimmunology ; 12(1): 2279800, 2023.
Article in English | MEDLINE | ID: mdl-38264343

ABSTRACT

Background: Non-responsiveness is a major barrier in current cancer immune checkpoint blockade therapies, and the mechanism has not been elucidated yet. Therefore, it is necessary to discover the mechanism and biomarkers of tumor immunotherapeutic resistance. Methods: Bioinformatics analysis was performed based on CD8+ T cell infiltration in multiple tumor databases to screen out genes related to anti-tumor immunity. Associations between Regulator of G-protein signaling 1 (RGS1) and IFNγ-STAT1 signaling, and MHCI antigen presentation pathway were examined by RT-qPCR, western blotting, and flow cytometry. The modulatory mechanisms of RGS1 were investigated via CHIP-qPCR and dual-luciferase assay. The clinical and therapeutic implications of RGS1 were comprehensively investigated using tumor cell lines, mouse models, and clinical samples receiving immunotherapy. Results: RGS1 was identified as the highest gene positively correlated with immunogenicity among RGS family. Inhibition of RGS1 in neoplastic cells dampened anti-tumor immune response and elicited resistance to immunotherapy in both renal and lung murine subcutaneous tumors. Mechanistically, RGS1 enhanced the binding of activating transcription factor 3 (ATF3) to the promoter of interferon gamma receptor 1 (IFNGR1), activated STAT1 and the subsequent expression of IFNγ-inducible genes, especially CXCL9 and MHC class I (MHCI), thereby influenced CD8+ T cell infiltration and antigen presentation and processing. Clinically, lower expression level of RGS1 was associated with resistance of PD1 inhibition therapy and shortened progression-free survival among 21 NSCLC patients receiving immunotherapy. Conclusions: Together, these findings uncover a novel mechanism that elicits immunotherapy resistance and highlight the function of tumor-intrinsic RGS1, which brings new insights for future strategies to sensitize anti-PD1 immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , RGS Proteins , Humans , Animals , Mice , Activating Transcription Factor 3 , Immunotherapy , Antigen Presentation
11.
Cell Death Discov ; 8(1): 243, 2022 May 03.
Article in English | MEDLINE | ID: mdl-35504887

ABSTRACT

Lymph node (LN) metastasis is associated with unfavorable prognosis of bladder cancer (BCa). Although lymphangiogenesis is functionally important in LN metastasis of tumors, the potential mechanism in BCa remains unclear. Here, we clarified a regulatory mechanism of circRNA-mediated lymphangiogenesis and LN metastasis in BCa based on next-generation sequencing data. We revealed that circDHTKD1 was positively associated with LN metastasis and significantly upregulated in BCa. By analyzing the co-expression patterns of circDHTKD1 and differentially expressed mRNAs, we identified that circDHTKD1 facilitated lymphangiogenesis by upregulating CXCL5. Mechanistically, circDHTKD1 directly interacted with miR-149-5p, and antagonized the repression of miR-149-5p on CXCL5. Furthermore, circDHTKD1-induced CXCL5 expression recruited and activated neutrophils, which participated in lymphangiogenesis by secreting VEGF-C. Our study supports circDHTKD1 as a promising diagnostic and therapeutic target for LN metastasis in BCa.

12.
J Hematol Oncol ; 15(1): 93, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35836291

ABSTRACT

BACKGROUND: Bladder cancer is the most common malignant tumor of the urinary system. Surgical resection and chemotherapy are the two mainstream treatments for bladder cancer. However, the outcomes are not satisfactory for patients with advanced bladder cancer. There is a need to further explore more effective targeted therapeutic strategies. METHODS: Proteomics were performed to compare protein expression differences between human bladder cancer tissues and adjacent normal tissues. The function of GPD1 on bladder cancer cells were confirmed through in vivo and in vitro assays. Transcriptomics and metabolomics were performed to reveal the underlying mechanisms of GPD1. Virtual screening was used to identify allosteric activator of GPD1. RESULTS: Here, we used proteomics to find that GPD1 expression was at low levels in bladder cancer tissues. Further investigation showed that GPD1 overexpression significantly promoted apoptosis in bladder cancer cells. Based on transcriptomics and metabolomics, GPD1 promotes Ca2+ influx and apoptosis of tumor cells via the lysoPC-PAFR-TRPV2 axis. Finally, we performed a virtual screening to obtain the GPD1 allosteric activator wedelolactone and demonstrated its ability to inhibit bladder tumor growth in vitro and in vivo. CONCLUSIONS: This study suggests that GPD1 may act as a novel tumor suppressor in bladder cancer. Pharmacological activation of GPD1 is a potential therapeutic approach for bladder cancer.


Subject(s)
Urinary Bladder Neoplasms , Allosteric Regulation , Apoptosis , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , TRPV Cation Channels/metabolism , Urinary Bladder Neoplasms/pathology
13.
Cell Death Discov ; 8(1): 439, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36319622

ABSTRACT

The mechanisms underlying the effects of cancer-associated fibroblasts (CAFs) on cancer stemness and tumor progression in renal cell carcinoma (RCC) have not been elucidated yet. In the present study, we found that the enrichment of CAFs was positively associated with tumor progression and cancer stemness in RCC. Further investigation revealed that CAFs could enhance cancer stemness through delivering exosomes to RCC cells, and miR-181d-5p was identified as the critical exosomal miRNA in CAF-secreted exosomes by small RNA sequencing and subsequent screening assays. Mechanistically, exosomal miR-181d-5p transferred from CAFs to RCC cells directly suppressed the expression of ring finger protein 43 (RNF43) and activated Wnt/ß-catenin signaling pathway, thus promoted cancer stemness and tumor progression. Overexpression of RNF43 strongly suppressed stemness properties and the effects could be reverted by miR-181d-5p. Overall, our findings revealed a crucial mechanism by which CAF-secreted exosomal miRNAs to enhance cancer stemness and thus promote RCC progression, suggesting a new avenue based on CAF-secreted miRNAs for more effective targeted therapies.

14.
Nat Commun ; 13(1): 4141, 2022 07 16.
Article in English | MEDLINE | ID: mdl-35842443

ABSTRACT

Organotropism during cancer metastasis occurs frequently but the underlying mechanism remains poorly understood. Here, we show that lysosomal protein transmembrane 5 (LAPTM5) promotes lung-specific metastasis in renal cancer. LAPTM5 sustains self-renewal and cancer stem cell-like traits of renal cancer cells by blocking the function of lung-derived bone morphogenetic proteins (BMPs). Mechanistic investigations showed that LAPTM5 recruits WWP2, which binds to the BMP receptor BMPR1A and mediates its lysosomal sorting, ubiquitination and ultimate degradation. BMPR1A expression was restored by the lysosomal inhibitor chloroquine. LAPTM5 expression could also serve as an independent predictor of lung metastasis in renal cancer. Lastly, elevation of LAPTM5 expression in lung metastases is a common phenomenon in multiple cancer types. Our results reveal a molecular mechanism underlying lung-specific metastasis and identify LAPTM5 as a potential therapeutic target for cancers with lung metastasis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I , Kidney Neoplasms , Lung Neoplasms , Ubiquitin-Protein Ligases , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Humans , Kidney Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
15.
Cancer Lett ; 522: 1-13, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34520818

ABSTRACT

The metastatic dissemination and underlying mechanisms of clear cell renal cell carcinoma (ccRCC) remain insufficiently understood. In this study, we identified the essential role of KLF2 in suppressing the metastasis of ccRCC. Downregulation of KLF2 detected by immunohistochemistry in primary metastatic ccRCC was remarkably related to poor clinical outcomes. Overexpression of KLF2 in vitro inhibited growth, migration and invasion of RCC cells. Analysis of clinical specimens revealed that there is a close correlation between KLF2 and GPX4 in ccRCC. Mechanistically, KLF2 deficiency is sufficient to inhibit ferroptosis on account of the impairment of transcriptional repression of GPX4 and thus promotes the migration and invasion of RCC cells. Reverting KLF2 expression in vivo decreased pulmonary metastatic lesions and prolonged life span of mice, whereas GPX4 overexpression reversed these properties. Overall, our results established a novel critical pathway that drives human ccRCC invasion and metastasis, which could be a promising target regarding to the therapies of advanced ccRCC in the clinic.


Subject(s)
Carcinoma, Renal Cell/genetics , Ferroptosis/genetics , Kruppel-Like Transcription Factors/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Aged , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Prognosis
16.
FEBS J ; 288(18): 5406-5429, 2021 09.
Article in English | MEDLINE | ID: mdl-33705609

ABSTRACT

Transcription factor SOX9 was a biomarker for prostate cancer (Pca) with poor prognosis. Nevertheless, the regulatory mechanism underlying SOX9 upregulation still remains unclear. Several cytokines have been reported to be involved in the regulation of SOX9, suggesting that cancer-associated fibroblasts (CAFs), one of the main sources of secreted factors in the tumor microenvironment (TME), may play a role in regulating SOX9 expression. Herein, an in vitro model of paracrine interaction between primary CAFs and Pca cells was applied to investigate the molecular mechanism of SOX9 upregulation during Pca progression. The regulatory axis was validated by in vivo experiments and The Cancer Genome Atlas data. Conditional medium of CAFs (CAF-CM) upregulated the expression of SOX9, which was mutually proved to be essential for CAF-induced tumor progression. Further analysis showed that hepatocyte growth factor (HGF) secreted by CAFs was responsible for SOX9 elevation in Pca cells, via the activation of c-Met signaling. Mechanistically, HGF/c-Met signaling specifically activated MEK1/2-ERK1/2 pathway, which induced phosphorylation and upregulation of FRA1, which then transcriptionally upregulated SOX9 by binding to the promoter of SOX9 gene. Moreover, we identified that HGF/c-Met-ERK1/2-FRA1-SOX9 axis was relatively conserved between human and mouse species by validating in mouse Pca cells. Our results reveal a novel insight into the molecular mechanism that SOX9 in Pca cells is promoted by CAFs through HGF/c-Met-ERK1/2-FRA1 axis. Furthermore, SOX9 may serve as an alternative marker for the activated HGF/c-Met signaling to enroll the optimal Pca patients for HGF/c-Met inhibition treatment, since it is much more stable and easier to detect.


Subject(s)
Hepatocyte Growth Factor/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-met/genetics , SOX9 Transcription Factor/genetics , Aged , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Female , Heterografts , Humans , MAP Kinase Signaling System/genetics , Male , Mice , Middle Aged , Paracrine Communication/genetics , Transcriptional Activation/genetics , Tumor Microenvironment/genetics
17.
Theranostics ; 10(17): 7683-7696, 2020.
Article in English | MEDLINE | ID: mdl-32685013

ABSTRACT

Tumor hypoxia, acidosis, and excessive reactive oxygen species (ROS) were the main characteristics of the bladder tumor microenvironment (TME), and abnormal TME led to autophagy activation, which facilitated cancer cell proliferation. The therapeutic efficacy of autophagy inhibitors might also be impeded by abnormal TME. To address these issues, we proposed a new strategy that utilized manganese dioxide (MnO2) nanoparticles to optimize the abnormal TME and revitalize autophagy inhibitors, and both oxygenation and autophagy inhibition may sensitize the tumor cells to radiation therapy. Methods: By taking advantage of the strong affinity between negatively charged MnO2 and positively charged chloroquine (CQ), the nanoparticles were fabricated by integrating MnO2 and CQ in human serum albumin (HSA)-based nanoplatform (HSA-MnO2-CQ NPs). Results: HSA-MnO2-CQ NPs NPs efficiently generated O2 and increased pH in vitro after reaction with H+/H2O2 and then released the encapsulated CQ in a H+/H2O2 concentration-dependent manner. The NPs restored the autophagy-inhibiting activity of chloroquine in acidic conditions by increasing its intracellular uptake, and markedly blocked hypoxia-induced autophagic flux. In vivo studies showed the NPs improved pharmacokinetic behavior of chloroquine and effectively accumulated in tumor tissues. The NPs exhibited significantly decreased tumor hypoxia areas and increased tumor pH, and had remarkable autophagy inhibition efficacy on bladder tumors. Finally, a significant anti-tumor effect achieved by the enhanced autophagy inhibition and radiation sensitization. Conclusions: HSA-MnO2-CQ NPs synergistically regulated the abnormal TME and inhibited autophagic flux, and effectively sensitized radiation therapy to treat bladder cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Chemoradiotherapy/methods , Drug Carriers/chemistry , Radiation-Sensitizing Agents/administration & dosage , Urinary Bladder Neoplasms/therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Autophagy/drug effects , Autophagy/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Chloroquine/administration & dosage , Chloroquine/pharmacokinetics , Drug Synergism , Humans , Hydrogen-Ion Concentration/drug effects , Male , Manganese Compounds/administration & dosage , Manganese Compounds/pharmacokinetics , Mice , Nanoparticles/chemistry , Oxides/administration & dosage , Oxides/pharmacokinetics , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacokinetics , Reactive Oxygen Species/metabolism , Serum Albumin, Human/chemistry , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
18.
ACS Appl Mater Interfaces ; 12(28): 31745-31756, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32571010

ABSTRACT

As a reactive oxygen species (ROS)-promoted disease, acute kidney injury (AKI) is associated with high mortality and morbidity, but no effective pharmacological treatment is available. Kidney-targeted and ROS-reactive antioxidants are in urgent demand for AKI treatment. A promising nanotechnology-based strategy for targeting renal tubules offers new perspectives for AKI treatment but remains challenging because of the glomerular filtration barrier, which requires ultrasmall-sized therapeutics for penetration and filtration. Here, we fabricated four potential antioxidative carbon nanodots (CNDs) with ultrasmall size. After balancing the antioxidant properties and biocompatibility, m-phenylenediamine-based CNDs (PDA-CNDs) were chosen for further research. PDA-CNDs demonstrated remarkable antioxidant properties for scavenging multiple toxic free radicals, enabling efficient protection of cells under various oxidative stresses in vitro. Moreover, fluorescence imaging revealed that PDA-CNDs preferentially accumulated in the injured kidney of mice with ischemia-reperfusion (IR)-induced AKI. Blood renal function tests and kidney tissue staining revealed the therapeutic efficacy of PDA-CNDs for AKI in both the murine IR-induced AKI model and cisplatin-induced AKI model. Collectively, this is the first study revealing that specific rationally designed CNDs could be a promising pharmacological treatment for AKI induced by ROS.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Carbon/chemistry , Phenylenediamines/chemistry , Animals , Antioxidants/metabolism , Cisplatin/adverse effects , Cisplatin/therapeutic use , Kidney/drug effects , Kidney/metabolism , Mice , Nanotechnology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism
19.
Biochim Biophys Acta Gen Subj ; 1863(9): 1360-1370, 2019 09.
Article in English | MEDLINE | ID: mdl-31170498

ABSTRACT

BACKGROUND: Renal fibrosis is a common pathological hallmark of chronic kidney disease, and no effective treatment is clinically available to manage its progression. Astaxanthin was recently found to be anti-fibrotic, but its effect on renal fibrosis remains unclear. METHODS: C57BL/6J mice were subjected to unilateral ureteral obstruction and intragastrically administered astaxanthin. Histopathology and immunohistochemistry were performed to evaluate renal fibrosis. Flow cytometry was used to examine lymphocyte accumulation in the fibrotic kidneys. Western blotting, real-time qPCR, and immunofluorescence were performed to cover the underlying mechanism concerning astaxanthin treatment during renal fibrosis. RESULTS: Oral administration of astaxanthin effectively alleviates renal fibrosis in mice. In vitro, astaxanthin inhibited fibroblast activation by modulating Smad2, Akt and STAT3 pathways and suppressed epithelial-to-mesenchymal transition in renal tubular epithelial cells through Smad2, snail, and ß-catenin. Moreover, astaxanthin significantly induced the rapid accumulation of CD8+ T cells in fibrotic kidneys, which was accompanied by elevated expression of IFN-γ. Accordingly, the depletion of CD8+ T cells strongly diminished the protective effect of astaxanthin. Further investigation showed that astaxanthin increased the population of CD8+ T cells by upregulating the expression of CCL5 in macrophages. CONCLUSIONS: These findings highlight the beneficial effect of astaxanthin on fibroblast activation, epithelial-to-mesenchymal transition, and CD8+ T cell recruitment during renal fibrosis. GENERAL SIGNIFICANCE: These data indicate that astaxanthin could serve as a therapeutic strategy to treat renal fibrotic conditions.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Fibrosis/prevention & control , Kidney Diseases/prevention & control , Myofibroblasts/cytology , Animals , CD8-Positive T-Lymphocytes/metabolism , Epithelial-Mesenchymal Transition/drug effects , Fibrosis/metabolism , Fibrosis/pathology , Interferon-gamma/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Smad2 Protein/metabolism , Snail Family Transcription Factors/metabolism , Xanthophylls/pharmacology , beta Catenin/metabolism
20.
Cancer Lett ; 449: 76-86, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30772441

ABSTRACT

Studies on the mechanism of clear cell renal cell carcinoma (ccRCC) progression are lacking. In this study, TOX3 was identified as a novel cancer suppressor gene in ccRCC. Hypermethylation of CpG probes in the promoter region was associated with the functional loss of TOX3 in ccRCC cancer tissues. Downregulation of TOX3 mRNA was strongly associated with poor clinical outcomes in ccRCC. Immunohistochemistry confirmed TOX3 was downregulated in primary tumors without metastasis (n = 126) and further downregulated in primary metastatic tumors (n = 23) compared with adjacent noncancerous tissues (n = 92). In vitro, overexpression of TOX3 inhibited RCC cell growth, migration and invasion. Mechanistic investigations showed that TOX3 deficiency facilitates the epithelial-mesenchymal transition due to impairment of transcriptional repression of SNAIL members SNAI1 and SNAI2 and promotes cancer cell migration and invasion. In vivo, restoring TOX3 expression reduced lung metastatic lesions and prolonged survival of mice. TOX3 combined with SNAI1 or SNAI2 predicted overall survival in ccRCC patients. Blockage of this pathway could be a promising therapeutic target for advanced ccRCC.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Snail Family Transcription Factors/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA Methylation , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Male , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Prognosis , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL