Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Mol Cell Probes ; 68: 101901, 2023 04.
Article in English | MEDLINE | ID: mdl-36791996

ABSTRACT

Gleason Score (GS) 3 + 4 prostate cancer (PCa) is heterogeneous in clinical course and molecular features. Risk stratification of indolent and aggressive PCa with GS 3 + 4 is critical, especially those with bone metastasis (BM) potential. Microarray-based microRNA(miRNA) profiling with eight PCa cases with or without BM was used to screen the candidate miRNAs associated with BM. Transwell and MTS assays were used to characterize the function of miRNAs and target gene LASP1. RT-qPCR and immunohistochemistry assays were utilized to illustrate the clinical significance of miRNAs and target gene in a cohort of 309 Chinese PCa cases. In the current study, we identified that miR-1-3p, miR-143-3p and miR-145-5p are associated with BM of GS 3 + 4 PCa. Through functional experiments, we show that miR-1-3p/143-3p/145-5p promotes proliferation and migration of PCa in vitro. LASP1 was predicted as the common target of these three miRNAs which was further confirmed by a luciferase assay. Overexpression of LASP1 was correlated with higher GS, higher pathological stage, and the presence of metastasis by immunohistochemistry. siRNA knockdown of LASP1 significantly suppressed proliferation and migration, whereas overexpression of LASP1 promoted it. Bioinformatics analysis revealed the involvement of Wnt signaling pathway in LASP1 mediated function. LASP1 may activate Wnt signaling by interacting with ß-catenin. In all, we suggest that miR-1-3p/143-3p/145-5p are associated with BM of Gleason 3 + 4 PCa. LASP1 is the common target of these miRNAs and may active Wnt signaling by interacting with ß-catenin.


Subject(s)
MicroRNAs , Prostatic Neoplasms , Male , Humans , beta Catenin/genetics , beta Catenin/metabolism , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , LIM Domain Proteins/genetics
2.
Biochem Biophys Res Commun ; 537: 22-28, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33383560

ABSTRACT

Triple-negative breast cancer (TNBC) is a major challenge in clinical practice due to its aggressiveness and lack of targeted treatment. Cancer stem-like traits contribute to tumorigenesis and immune privilege of TNBC. However, the relationship of stemness and immunosurveillance remains unclear. Here, we demonstrate that BTF3 expression is related with stem-like properties in TNBC cells. BTF3 modulates stemness, migration and proliferation of TNBC in vitro. Bioinformatics analysis revealed that interferon signaling pathways and IRF7, both of which participate in the immune escape of TNBC, are closely related to BTF3 in TNBC cells. Knockdown of BTF3 activates IRF7 expression through increased degradation of BMI1, a protein that can represses IRF7 transcription by directly binding to its promotor region. BTF3 links stem-like traits and the interferon signaling pathway, revealing the potential connection of stemness and immunomodulation in TNBC. Clinically, we suggest that BTF3 is predictive of poor prognosis in patients with TNBC. Together, our findings highlight an important role of BTF3 in regulating the progression of TNBC cells.


Subject(s)
Interferon Type I/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Polycomb Repressive Complex 1/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Treatment Outcome , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
3.
Prostate ; 80(9): 674-686, 2020 06.
Article in English | MEDLINE | ID: mdl-32294305

ABSTRACT

BACKGROUND: Castrate-resistant prostate cancer (CRPC) is an aggressive and lethal disease. The pathogenesis of CRPC is not fully understood and novel therapeutic targets need to be identified to improve the patients' prognosis. MicroRNA-30a (miR-30a) has been demonstrated to be a tumor suppressor in many types of solid malignancies. However, its role in androgen-independent (AI) growth of prostate cancer (PCa) received limited attention as yet. METHODS: The clinical association of miR-30a and its potential targets with AI growth was characterized by bioinformatics analyses. Regulation of cell proliferation and colony formation rates by miR-30a were tested using PCa cell models. Xenograft models were used to measure the regulation of prostate tumor growth by miR-30a. The real-time quantitative polymerase chain reaction was used to validate whether miR-30a and its targets regulate cell cycle control genes and androgen receptor (AR)-dependent transcription. Bioinformatics tools, Western blot, and luciferase reporter assays were utilized to identify miR-30a targets. RESULTS: Bioinformatic analysis showed that low expression of miR-30a is associated with castration resistance of PCa patients and poor outcomes. Transfection of miR-30a mimics inhibited the AI growth of PCa cells in vitro and in vivo. Upregulation of miR-30a in 22RV1 cells altered the expression of cell cycle control genes and AR-mediated transcription, while downregulation of miR-30a in LNCaP cells had the opposite effects to AR-mediated transcription. MYBL2, FOXD1, and SOX4 were identified as miR-30a targets. Downregulation of MYBL2, FOXD1, and SOX4 affected the expression of cell cycle control genes and AR-mediated transcription and suppressed the AI growth of 22RV1 cells. CONCLUSIONS: Our results suggest that miR-30a inhibits AI growth of PCa by targeting MYBL2, FOXD1, and SOX4. They provide novel insights into developing new treatment strategies for CRPC.


Subject(s)
Cell Cycle Proteins/metabolism , Forkhead Transcription Factors/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , SOXC Transcription Factors/metabolism , Trans-Activators/metabolism , Androgen Antagonists/metabolism , Androgens/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Down-Regulation , Forkhead Transcription Factors/genetics , HEK293 Cells , Heterografts , Humans , Male , Mice , Mice, Nude , MicroRNAs/genetics , Prognosis , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/metabolism , SOXC Transcription Factors/genetics , Trans-Activators/genetics , Up-Regulation
4.
Angew Chem Int Ed Engl ; 57(51): 16716-16720, 2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30370977

ABSTRACT

Hydrothermal-synthesis-based reactions are normally single step owing to the difficulty of manipulating reaction mixtures at high temperatures and pressures. Herein we demonstrate a simple, cheap, and modular approach to the design reactors consisting of partitioned chambers, to achieve multi-step synthesis under hydrothermal conditions, in digitally defined reactionware produced by 3D printing. This approach increases the number of steps that can be performed sequentially and allows an increase in the options available for the control of hydrothermal reactions. The synthetic outcomes of the multi-stage reactions can be explored by varying reaction compositions, number of reagents, reaction steps, and reaction times, and these can be tagged to the digital blueprint. To demonstrate the potential of this approach a series of polyoxometalate (POM)-containing metal-organic frameworks (MOFs) unavailable by "one-pot" methods were prepared as well as a set of new MOFs.

5.
J Bone Miner Metab ; 34(5): 500-16, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26210858

ABSTRACT

Numerous studies have demonstrated that rheumatoid arthritis (RA) is often associated with bone loss; however, few experiments have focused on cancellous and cortical bone changes in rats during the process of arthritis. We have investigated bone changes in rats with collagen-induced arthritis (CIA) and have explored the characteristics of how RA induces osteoporosis by means of bone histomorphometry, bone biomechanics studies, bone mineral density studies, micro computer tomography, enzyme-linked immunosorbant assay, immunohistochemistry, and Western blot analysis. Bone mineral density of the femur and lumbar vertebrae and biomechanical properties of the femur were decreased in CIA rats. Trabecular bone volume of the tibia and lumbar vertebrae was decreased whereas bone resorption was increased in CIA rats. Bone formation of the tibial shaft in periosteal surfaces was decreased in CIA rats. Furthermore, the trabecular bone loss in CIA rats was severer at 16 weeks than at 8 weeks, as was cortical bone loss. The serum level of tumor necrosis factor α in CIA rats was increased, and the expression of dickkopf 1 and that of receptor activator of nuclear factor κB (RANKL) ligand (RANKL) in the ankle joints were also increased, but the expression of osteoprotegerin (OPG) was decreased. We conclude that CIA rats developed systemic osteoporosis, and that osteoporosis became more serious with CIA development. The mechanism may be related to the increase of bone resorption in cancellous bone cause by upregulation of the expression of DKK-1 and regulation of the RANKL/RANK/OPG signaling pathway, and the decrease of bone formation in cortical bone caused by an increase in the expression of DKK-1.


Subject(s)
Arthritis, Experimental/complications , Osteoporosis/etiology , Animals , Ankle Joint/pathology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Biomechanical Phenomena , Bone Density , Cancellous Bone/pathology , Female , Femur/physiopathology , Intercellular Signaling Peptides and Proteins/metabolism , Osteoporosis/metabolism , Osteoporosis/pathology , Osteoporosis/physiopathology , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Rats , Rats, Inbred Lew
6.
Huan Jing Ke Xue ; 44(2): 912-923, 2023 Feb 08.
Article in Zh | MEDLINE | ID: mdl-36775614

ABSTRACT

As the largest terrestrial carbon pool, the spatial distribution characteristics and influencing factors of soil organic carbon have important implications for global carbon cycle processes. Soil organic carbon density (SOCD) and influencing factors were predicted in the Yellow River basin using a mixed geographically weighted regression (MGWR) model based on soil organic carbon density data and environmental factors. The results showed that:① the SOCD ranged from 0-14.82 kg·m-2 and 0-32.39 kg·m-2 for the soil depths of 0-20 cm and 0-100 cm, with mean values of 3.48 kg·m-2 and 8.07 kg·m-2 and reserves of 2.76 Pg and 6.48 Pg, respectively. The high SOCD value areas were mainly located in the southern part of the Qinghai-Tibet Plateau and Loess Plateau, and the low value areas were located in the eastern part of the upper Yellow River and the inland flow area. ②Among the ecosystem types, the SOCD of soil depth in 0-20 cm was in the descending order of:forest>water body and wetland>other>grassland>farmland>settlement>desert, with mean values of 4.52, 4.31, 3.84, 3.73, 2.89, 2.78, and 2.22 kg·m-2, respectively, and the SOCD of the 0-100 cm soil depth was in the descending order of:water bodies and wetlands>forest>other>grassland>farmland>settlement>desert, with mean values of 9.58, 9.58, 8.85, 8.66, 7.07, 6.81, and 5.29 kg·m-2, respectively. The SOCR in descending order was:grassland>farmland>forest>desert>water bodies and wetlands>settlement>others, with 1.40, 0.60, 0.47, 0.11, 0.07, 0.06, and 0.05 Pg at a soil depth of 0-20 cm and 3.31, 1.49, 0.99, 0.26, 0.17, 0.14, and 0.12 Pg at a soil depth of 0-100 cm, respectively. ③ The main factors affecting the SOCD distribution were intercept, profile curvature, NDVI, and precipitation; in addition, curvature and silt also had important effects on the deep SOCD distribution in the Yellow River basin. Among the ecosystem types, precipitation and NDVI were the main factors affecting the SOCD distribution. The intercept also had important effects on the SOCD distribution in the all ecosystems except forests, whereas curvature and silt only had important effects on deserts and other ecosystems. These results revealed the spatial distribution of SOCD, influencing factors, and SOCR in the Yellow River basin and can provide a scientific basis for carbon balance, soil quality evaluation, and ecological management restoration and consolidation in the region.

7.
bioRxiv ; 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37732209

ABSTRACT

Widespread sequencing has yielded thousands of missense variants predicted or confirmed as disease-causing. This creates a new bottleneck: determining the functional impact of each variant - largely a painstaking, customized process undertaken one or a few genes or variants at a time. Here, we established a high-throughput imaging platform to assay the impact of coding variation on protein localization, evaluating 3,547 missense variants of over 1,000 genes and phenotypes. We discovered that mislocalization is a common consequence of coding variation, affecting about one-sixth of all pathogenic missense variants, all cellular compartments, and recessive and dominant disorders alike. Mislocalization is primarily driven by effects on protein stability and membrane insertion rather than disruptions of trafficking signals or specific interactions. Furthermore, mislocalization patterns help explain pleiotropy and disease severity and provide insights on variants of unknown significance. Our publicly available resource will likely accelerate the understanding of coding variation in human diseases.

8.
J Exp Clin Cancer Res ; 41(1): 125, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35382861

ABSTRACT

BACKGROUND: The development of castration-resistant prostate cancer (CRPC) remains a major obstacle in the treatment of prostate cancer (PCa). Dysregulated mitochondrial function has been linked to the initiation and progression of diverse human cancers. Deciphering the novel molecular mechanisms underlying mitochondrial function may provide important insights for developing novel therapeutics for CRPC. METHODS: We investigate the expression of the protein tyrosine phosphatase receptor type F polypeptide interacting protein alpha 4 (PPFIA4) using public datasets and tumor specimens from PCa cases by immunohistochemistry. Gain- and loss-of-function studies are performed in PCa cell lines and mouse models of subcutaneous xenograft to characterize the role of PPFIA4 in CRPC. Gene expression regulation is evaluated by a series of molecular and biochemical experiments in PCa cell lines. The therapeutic effects of methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) inhibitor combined enzalutamide are assessed using in vitro functional assays and in vivo mouse models. RESULTS: We show that the increase of PPFIA4 exacerbates aggressive phenotype resembling CRPC. A fraction of PPFIA4 localizes to mitochondria and interacts with MTHFD2, a key enzyme for one-carbon metabolism. Androgen deprivation increases the translocation of PPFIA4 into mitochondria and increases the interaction between PPFIA4 and MTHFD2, which result in the elevation of tyrosine phosphorylated MTHFD2. Consequently, the levels of NADPH synthesis increase, resulting in protection against androgen deprivation-induced mitochondrial dysfunction, as well as promotion of tumor growth. Clinically, PPFIA4 expression is significantly increased in CRPC tissues compared with localized PCa ones. Importantly, an MTHFD2 inhibitor, DS18561882, combined with enzalutamide can significantly inhibit CRPC cell proliferation in vitro and tumor growth in vivo. CONCLUSION: Overall, our findings reveal a PPFIA4-MTHFD2 complex in mitochondria that links androgen deprivation to mitochondrial metabolism and mitochondrial dysfunction, which suggest a potential strategy to inhibit CRPC progression.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Androgen Antagonists , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mitochondria/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology
9.
Cancer Res ; 81(4): 1026-1039, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33277366

ABSTRACT

The new generation androgen receptor (AR) pathway inhibitor enzalutamide can prolong the survival of patients with metastatic prostate cancer. However, resistance to enzalutamide inevitably develops in these patients, and the underlying mechanisms of this resistance are not fully defined. Here we demonstrate that the kinesin family member 15 (KIF15) contributes to enzalutamide resistance by enhancing the AR signaling in prostate cancer cells. KIF15 directly bound the N-terminus of AR/AR-V7 and prevented AR/AR-V7 proteins from degradation by increasing the protein association of ubiquitin-specific protease 14 (USP14) with AR/AR-V7. In turn, the transcriptionally active AR stimulated KIF15 expression. KIF15 inhibitors alone or in combination with enzalutamide significantly suppressed enzalutamide-resistant prostate cancer cell growth and xenograft progression. These findings highlight a key role of KIF15 in enabling prostate cancer cells to develop therapy resistance to enzalutamide and rationalize KIF15 as a potential therapeutic target. SIGNIFICANCE: These findings demonstrate how reciprocal activation between KIF15 and AR contributes to enzalutamide resistance in prostate cancer and highlights cotargeting KIF15 and AR as a therapeutic strategy for these tumors.


Subject(s)
Benzamides/therapeutic use , Drug Resistance, Neoplasm/genetics , Kinesins/physiology , Nitriles/therapeutic use , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , Disease Progression , HEK293 Cells , Humans , Kinesins/genetics , Male , Mice , Mice, Nude , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Domains/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Stability , Proteolysis , Receptors, Androgen/chemistry , Receptors, Androgen/genetics
10.
Front Oncol ; 11: 679173, 2021.
Article in English | MEDLINE | ID: mdl-34804913

ABSTRACT

Castration-resistant prostate cancer (CRPC) continues to be a major clinical problem and its underlying mechanisms are still not fully understood. The epidermal growth factor receptor (EGFR) activation is an important event that regulates mitogenic signaling. EGFR signaling plays an important role in the transition from androgen dependence to castration-resistant state in prostate cancer (PCa). Kinesin family member 15 (KIF15) has been suggested to be overexpressed in multiple malignancies. Here, we demonstrate that KIF15 expression is elevated in CRPC. We show that KIF15 contributes to CRPC progression by enhancing the EGFR signaling pathway, which includes complex network intermediates such as mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. In CRPC tumors, increased expression of KIF15 is positively correlated with EGFR protein level. KIF15 binds to EGFR, and prevents EGFR proteins from degradation in a Cdc42-dependent manner. These findings highlight the key role of KIF15 in the development of CRPC and rationalize KIF15 as a potential therapeutic target.

11.
Oncogene ; 39(2): 356-367, 2020 01.
Article in English | MEDLINE | ID: mdl-31477836

ABSTRACT

Reactive oxygen species (ROS) and ROS-induced oxidative stress are associated with prostate cancer (PCa) development and castrate-resistant tumor progression. This is in part through the activation of the androgen receptor (AR) signaling. However, the molecular underpinning of ROS to activate AR remains poorly understood. Here, we report that the thioredoxin domain-containing 9 (TXNDC9) is an important regulator of ROS to trigger AR signaling. TXNDC9 expression is upregulated by ROS inducer, and increased TXNDC9 expression in patient tumors is associated with advanced clinical stages. TXNDC9 promotes PCa cell survival and proliferation. It is required for AR protein expression and AR transcriptional activity under oxidative stress conditions. Mechanistically, ROS inducers promote TXNDC9 to dissociate from PRDX1, but enhance a protein association with MDM2. Concurrently, PRDX1 enhances its association with AR. These protein interaction exchanges result in not only MDM2 protein degradation, but also PRDX1 mediated AR protein stabilization, and subsequent elevation of AR signaling. Blocking PRDX1 by its inhibitor, Conoidin A (CoA), suppresses AR signaling, PCa cell proliferation, and xenograft tumor growth even under androgen-deprived conditions. These tumor-suppressive effects of CoA were further strengthened when in combination with enzalutamide treatment. Together, these studies demonstrate that the TXNDC9-PRDX1 axis plays an important role for ROS to activate AR functions. It provides a proof-of-principle that co-targeting AR and PRDX1 may be more effective to control PCa growth.


Subject(s)
Peroxiredoxins/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/genetics , Thioredoxins/genetics , Animals , Benzamides , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Male , Mice , Nitriles , Oxidative Stress/genetics , Peroxiredoxins/antagonists & inhibitors , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Stability , Proto-Oncogene Proteins c-mdm2/genetics , Quinoxalines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction
12.
J Exp Clin Cancer Res ; 38(1): 227, 2019 May 28.
Article in English | MEDLINE | ID: mdl-31138311

ABSTRACT

BACKGROUND: Cancer stem-like traits contribute to prostate cancer (PCa) progression and metastasis. Deciphering the novel molecular mechanisms underlying stem-like traits may provide important insight for developing novel therapeutics. METHODS: Immunohistochemistry and immunofluorescence assays in prostatic tissues; gain- and loss-of-function analyses using ectopic overexpression and shRNAs in PCa cell lines; measurements of tumorigenic and stemness properties, and transcription in vitro and in vivo; transcriptional analysis in public databases. RESULTS: We identified that overexpression of BTF3 in PCa tissues and BTF3 expression highly correlates to stem-like traits. Cancer stem-like characteristics in PCa including self-renewal and metastatic potential were impaired by BTF3 loss and promoted by BTF3 overexpression. Mechanistically, BTF3 could stabilize BMI1, which is a crucial regulator of prostate stem cell self-renewal. More importantly, our data revealed that BTF3 is highly predictive of poor prognosis and may help in risk stratification of PCa patients. CONCLUSIONS: BTF3 promotes PCa progression though modeling stem-like traits in PCa. BTF3 represents a stratification marker in PCa progression and outcomes.


Subject(s)
Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1/chemistry , Prostatic Neoplasms/pathology , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Prostatic Neoplasms/metabolism , Protein Stability , Survival Analysis , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL