Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
bioRxiv ; 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-39026690

ABSTRACT

Noncoding RNAs (ncRNAs) are increasingly recognized as bioactive. Here we report the development of TY1, a synthetic ncRNA bioinspired by a naturally-occurring human small Y RNA with immunomodulatory properties. TY1 upregulates TREX1, an exonuclease that rapidly degrades cytosolic DNA. In preclinical models of myocardial infarction (MI) induced by ischemia/reperfusion, TY1 reduced scar size. The cardioprotective effect of TY1 was abrogated by prior depletion of macrophages and mimicked by adoptive transfer of macrophages exposed either to TY1 or TREX1. Inhibition of TREX1 in macrophages blocked TY1 cardioprotection. Consistent with a central role for TREX1, TY1 attenuated DNA damage in the post-MI heart. This novel mechanism-pharmacologic upregulation of TREX1 in macrophages-establishes TY1 as the prototype for a new class of ncRNA drugs with disease-modifying bioactivity. One Sentence Summary: Upregulation of three prime exonuclease, TREX1, in macrophages enhances tissue repair post myocardial infarction.

3.
bioRxiv ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38746093

ABSTRACT

All approved RNA therapeutics require parenteral delivery. Here we demonstrate an orally bioavailable formulation wherein synthetic noncoding (nc) RNA, packaged into lipid nanoparticles, is loaded into casein-chitosan (C2) micelles. We used the C2 formulation to deliver TY1, a 24-nucleotide synthetic ncRNA which targets the DNA damage response pathway in macrophages. C2-formulated TY1 (TY1C2) efficiently packages and protects TY1 against degradative enzymes. In healthy mice, oral TY1C2 was well-tolerated and nontoxic. Oral TY1C2 exhibited disease-modifying bioactivity in 2 models of tissue injury: 1) rat myocardial infarction, where a single oral dose of TY1C2 was cardioprotective, on par with intravenously-delivered TY1; and 2) mouse acute lung injury, where a single dose of TY1C2 attenuated pulmonary inflammation. Mechanistic dissection revealed that TY1C2 is not absorbed into the systemic circulation but is, instead, taken up by intestinal macrophages, namely those of the lamina propria and Peyer's patches. This route of absorption may rationalize why an antisense oligonucleotide against Factor VII, which acts on hepatocytes, is not effective when administered in the C2 formulation. Thus, some (but not all) ncRNA drugs are bioavailable when delivered by mouth. Oral RNA delivery and uptake, relying on uptake via the gastrointestinal immune system, has broad-ranging therapeutic implications.

4.
Mol Ther Nucleic Acids ; 24: 951-960, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34094713

ABSTRACT

Cardiosphere-derived cell exosomes (CDCexo) and YF1, a CDCexo-derived non-coding RNA, elicit therapeutic bioactivity in models of myocardial infarction and hypertensive hypertrophy. Here we tested the hypothesis that YF1, a 56-nucleotide Y RNA fragment, could alleviate cardiomyocyte hypertrophy, inflammation, and fibrosis associated with hypertrophic cardiomyopathy (HCM) in transgenic mice harboring a clinically relevant mutation in cardiac troponin I (cTnIGly146). By quantitative PCR, YF1 was detectable in bone marrow, spleen, liver, and heart 30 min after intravenous (i.v.) infusion. For efficacy studies, mice were randomly allocated to receive i.v. YF1 or vehicle, monitored for ambulatory and cardiac function, and sacrificed at 4 weeks. YF1 (but not vehicle) improved ambulation and reduced cardiac hypertrophy and fibrosis. In parallel, peripheral mobilization of neutrophils and proinflammatory monocytes was decreased, and fewer macrophages infiltrated the heart. RNA-sequencing of macrophages revealed that YF1 confers substantive and broad changes in gene expression, modulating pathways associated with immunological disease and inflammatory responses. Together, these data demonstrate that YF1 can reverse hypertrophic and fibrotic signaling pathways associated with HCM, while improving function, raising the prospect that YF1 may be a viable novel therapeutic candidate for HCM.

5.
J Vis Exp ; (147)2019 05 28.
Article in English | MEDLINE | ID: mdl-31205316

ABSTRACT

The beneficial effects of reperfusion therapy for ST-segment elevation myocardial infarction (STEMI) is attenuated by reperfusion injury. No approach has been proven successful in preventing this injury in the clinical setting to date. Meanwhile, a novel approach for cardioprotection in patients with STEMI, i.e., postconditioning with lactate-enriched blood (PCLeB), has recently been reported. PCLeB is a modification of the original protocol of postconditioning, aimed at increasing the delay in the recovery from tissue acidosis produced during ischemia. This was sought to achieve controlled reperfusion with tissue oxygenation and minimal lactate washout. In this modified postconditioning protocol, the duration of each brief reperfusion is gradually increased in a stepwise manner from 10 to 60 s. Each brief ischemic period lasts for 60 s. At the end of each brief reperfusion, injection of lactated Ringer's solution (20-30 mL) is performed directly into the culprit coronary artery immediately before the balloon inflation and the balloon is quickly inflated at the lesion site, so that the lactate is trapped inside the ischemic myocardium during each brief repetitive ischemic period. After seven cycles of balloon inflation and deflation, full reperfusion is performed. Stenting is performed thereafter, and the percutaneous coronary intervention is completed. Excellent in-hospital and 6 month outcomes in a limited number of patients with STEMI treated using PCLeB have already been reported. This method article provides a detailed description of each step of the PCLeB procedures.


Subject(s)
Ischemic Postconditioning/methods , Lactates/metabolism , Percutaneous Coronary Intervention/methods , ST Elevation Myocardial Infarction/therapy , Female , Humans , Male , ST Elevation Myocardial Infarction/pathology , Stents
6.
Int J Oncol ; 54(6): 2222-2236, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30942440

ABSTRACT

miRNA­223 (miR­223) has been reported to function not only as a tumor suppressor, but also as an oncogenic microRNA (miRNA or miR) in various cancer cells. Therefore, the functional role of miR­223 has not been elucidated to date, at least to the best of our knowledge. We previously performed the deep sequencing analysis of clinical bladder cancer (BC) specimens. It was revealed that miR­223 expression was significantly downregulated in BC, suggesting that miR­223 functions as a tumor suppressor miRNA in BC. The aim of this study was to investigate the functional roles of miR­223 and to identify its targets in BC. The expression levels of miR­223 were significantly decreased in our clinical BC specimens. The Cancer Genome Atlas (TCGA) database indicated that miR­223 expression was related to lymphovascular invasion and distant metastasis. The restoration of miR­223 expression significantly inhibited tumor aggressiveness and induced apoptosis via caspase­3/7 activation in BC cells. WD repeat domain 62 (WDR62), a candidate target of miR­223 according to in silico analyses, has been previously proposed to play a role in neurodevelopment. Direct binding between WDR62 and miR­223 was confirmed by luciferase assay. The TCGA database revealed positive associations between WDR62 mRNA expression and a higher tumor grade and stage in BC. The knockdown of WDR62 significantly inhibited tumor aggressiveness and induced the apoptosis of BC cells. On the whole, the findings of this study reveal a novel miR­223 target, oncogenic WDR62, and provided insight into the oncogenesis of BC.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Nerve Tissue Proteins/genetics , Oncogene Proteins/genetics , Urinary Bladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cystectomy , Datasets as Topic , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Neoplasm Grading , Urinary Bladder/pathology , Urinary Bladder/surgery , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery
7.
Nat Commun ; 10(1): 674, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30787297

ABSTRACT

Direct cardiac reprogramming from fibroblasts can be a promising approach for disease modeling, drug screening, and cardiac regeneration in pediatric and adult patients. However, postnatal and adult fibroblasts are less efficient for reprogramming compared with embryonic fibroblasts, and barriers to cardiac reprogramming associated with aging remain undetermined. In this study, we screened 8400 chemical compounds and found that diclofenac sodium (diclofenac), a non-steroidal anti-inflammatory drug, greatly enhanced cardiac reprogramming in combination with Gata4, Mef2c, and Tbx5 (GMT) or GMT plus Hand2. Intriguingly, diclofenac promoted cardiac reprogramming in mouse postnatal and adult tail-tip fibroblasts (TTFs), but not in mouse embryonic fibroblasts (MEFs). Mechanistically, diclofenac enhanced cardiac reprogramming by inhibiting cyclooxygenase-2, prostaglandin E2/prostaglandin E receptor 4, cyclic AMP/protein kinase A, and interleukin 1ß signaling and by silencing inflammatory and fibroblast programs, which were activated in postnatal and adult TTFs. Thus, anti-inflammation represents a new target for cardiac reprogramming associated with aging.


Subject(s)
Cellular Reprogramming/drug effects , Cyclooxygenase 2/pharmacology , Myocytes, Cardiac/drug effects , Receptors, Prostaglandin E, EP4 Subtype/drug effects , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cyclic AMP , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclooxygenase 2/drug effects , Diclofenac/pharmacology , Dinoprostone , Fibroblasts , GATA4 Transcription Factor/metabolism , Humans , Inflammation , Interleukin-1beta , MEF2 Transcription Factors/metabolism , Mice , Mice, Transgenic , T-Box Domain Proteins/metabolism
8.
Int J Cardiol ; 275: 36-38, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30309681

ABSTRACT

BACKGROUND: We recently reported a new approach for cardioprotection, postconditioning with lactate-enriched blood (PCLeB), and a patient with ST-segment elevation myocardial infarction (STEMI), in whom muscle squeezing of the culprit coronary artery was observed immediately after reperfusion with PCLeB. In this study, we examined the prevalence of muscle squeezing immediately after reperfusion in patients with anterior STEMI treated using PCLeB. METHODS AND RESULTS: PCLeB is a modified postconditioning protocol that comprises intermittent reperfusion and timely coronary injections of lactated Ringer's solution. We treated 30 consecutive patients with anterior STEMI using PCLeB. Among the 30 patients, 4 patients exhibited muscle squeezing of the left anterior descending artery (LAD) immediately after reperfusion. We performed follow-up coronary angiography in 23 patients and found another patient who exhibited muscle squeezing of the LAD. Thus, of 30 patients, 5 were confirmed to have myocardial bridging and 4 exhibited muscle squeezing immediately after reperfusion with PCLeB. No patient died or experienced re-hospitalization for heart failure or recurrent ischemic events at 6 months except for one patient with malignancy. CONCLUSION: Muscle squeezing immediately after reperfusion therapy is not a rare phenomenon in patients with anterior STEMI treated using PCLeB.


Subject(s)
Coronary Circulation/drug effects , Coronary Vessels/physiopathology , Ischemic Preconditioning, Myocardial/methods , Ringer's Lactate/administration & dosage , ST Elevation Myocardial Infarction/therapy , Coronary Vessels/drug effects , Electrocardiography , Female , Follow-Up Studies , Humans , Male , Middle Aged , Retrospective Studies , ST Elevation Myocardial Infarction/diagnosis , Time Factors
9.
Int J Oncol ; 53(2): 725-736, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29901113

ABSTRACT

The active form of the small GTPase RAS binds to downstream effectors to promote cell growth and proliferation. RAS signal enhancement contributes to tumorigenesis, invasion, and metastasis in various different cancers. HRAS proto-oncogene GTPase (HRAS), one of the RAS isoforms, was the first human oncogene for which mutations were reported in T24 bladder cancer (BC) cells in 1982, and HRAS mutation or upregulation has been reported in several cancers. According to data from The Cancer Genome Atlas, HRAS expression was significantly upregulated in clinical BC samples compared to healthy samples (P=0.0024). HRAS expression was also significantly upregulated in BC with HRAS mutation compared to patients without HRAS mutation (P<0.0001). The tumor suppressive effect of salirasib, a RAS inhibitor, has been reported in several cancer types, but only at relatively high concentrations. As such, RAS inhibitors have not been used for clinical applications. The aim of the current study was to investigate the therapeutic potential of targeting HRAS using salirasib and small interfering RNA (siRNA) and to characterize the mechanism by which HRAS functions using recently developed quantitative in vitro proteome-assisted multiple reaction monitoring for protein absolute quantification (iMPAQT), in BC cells. iMPAQT allows measurement of the absolute abundance of any human protein with the high quantitative accuracy. Salirasib and siRNA targeting of HRAS inhibited cell proliferation, migration and invasion in HRAS wild type and HRAS-mutated cell lines. Proteomic analyses revealed that several metabolic pathways, including the oxidative phosphorylation pathway and glycolysis, were significantly downregulated in salirasib-treated BC cells. However, the expression levels of hexokinase 2, phosphoglycerate kinase 1, pyruvate kinase, muscle (PKM)1, PKM2 and lactate dehydrogenase A, which are downstream of RAS and target genes of hypoxia inducible factor-1α, were not notably downregulated, which may explain the high concentration of salirasib required to inhibit cell viability. These findings provide insight into the mechanisms of salirasib, and suggest the need for novel therapeutic strategies to treat cancers such as BC.


Subject(s)
Antineoplastic Agents/administration & dosage , Farnesol/analogs & derivatives , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Salicylates/administration & dosage , Up-Regulation/drug effects , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Farnesol/administration & dosage , Farnesol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Regulatory Networks , Glycolysis/drug effects , Humans , Mice , Mutation , Oxidative Phosphorylation/drug effects , Proteomics , Proto-Oncogene Mas , Salicylates/pharmacology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Xenograft Model Antitumor Assays
10.
Oncotarget ; 9(33): 23003-23017, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29796168

ABSTRACT

Sunitinib is a standard molecular-targeted drug used as a first-line treatment for metastatic clear cell renal cell carcinoma (ccRCC); however, resistance to sunitinib has become a major problem in medical practice. Recently, bromodomain containing 4 (BRD4), a member of the bromodomain family proteins, was identified as a promising therapeutic target, and its inhibitor JQ1 has been shown to have inhibitory effects in various human cancers. However, the anti-cancer effects of JQ1 in ccRCC, particularly sunitinib-resistant ccRCC, are still unclear. Here, we aimed to elucidate the anti-cancer effects of JQ1 and the mechanisms underlying BRD4 inhibition in sunitinib-sensitive and -resistant ccRCCs. Analysis of The Cancer Genome Atlas (TCGA) ccRCC cohort showed that patients with high BRD4 expression had shorter overall survival than those with low expression. JQ1 treatment significantly inhibited tumor growth of sunitinib-sensitive and -resistant ccRCC cells in part through MYC regulation. Based on RNA sequencing analyses of ccRCC cells treated with JQ1 to elucidate the mechanisms other than MYC regulation, we identified several oncogenes that may be potential therapeutic targets or prognostic markers; patients with high expression of SCG5, SPOCD1, RGS19, and ARHGAP22 had poorer overall survival than those with low expression in TCGA ccRCC cohort. Chromatin immunoprecipitation assays revealed that these oncogenes may be promising BRD4 targets, particularly in sunitinib-resistant ccRCC cells. These results identified SCG5, SPOCD1, RGS19, and ARHGAP22 as potential prognostic markers and showed that BRD4 inhibition may have applications as a potential therapeutic approach in sunitinib-sensitive and -resistant ccRCC.

12.
J Am Heart Assoc ; 7(1)2018 01 07.
Article in English | MEDLINE | ID: mdl-29307865

ABSTRACT

BACKGROUND: It has never been possible to immediately evaluate heart rate variability (HRV) during exercise. We aimed to visualize the real-time changes in the power spectrum of HRV during exercise and to investigate its relationship to the ventilatory threshold (VT). METHODS AND RESULTS: Thirty healthy subjects (29.1±5.7 years of age) and 35 consecutive patients (59.0±13.2 years of age) with myocardial infarctions underwent cardiopulmonary exercise tests with an RAMP protocol ergometer. The HRV was continuously assessed with power spectral analyses using the maximum entropy method and projected on a screen without delay. During exercise, a significant decrease in the high frequency (HF) was followed by a drastic shift in the power spectrum of the HRV with a periodic augmentation in the low frequency/HF (L/H) and steady low HF. When the HRV threshold (HRVT) was defined as conversion from a predominant high frequency (HF) to a predominant low frequency/HF (L/H), the VO2 at the HRVT (HRVT-VO2) was substantially correlated with the VO2 at the lactate threshold and VT) in the healthy subjects (r=0.853 and 0.921, respectively). The mean difference between each threshold (0.65 mL/kg per minute for lactate threshold and HRVT, 0.53 mL/kg per minute for VT and HRVT) was nonsignificant (P>0.05). Furthermore, the HRVT-VO2 was also correlated with the VT-VO2 in these myocardial infarction patients (r=0.867), and the mean difference was -0.72 mL/kg per minute and was nonsignificant (P>0.05). CONCLUSIONS: A HRV analysis with our method enabled real-time visualization of the changes in the power spectrum during exercise. This can provide additional information for detecting the VT.


Subject(s)
Anaerobic Threshold , Cardiorespiratory Fitness , Electrocardiography/methods , Exercise Test/methods , Exercise Tolerance , Heart Rate , Lung/physiopathology , Myocardial Infarction/diagnosis , Signal Processing, Computer-Assisted , Adult , Aged , Bicycling , Biomarkers/blood , Case-Control Studies , Female , Humans , Lactic Acid/blood , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/physiopathology , Predictive Value of Tests , Reproducibility of Results , Respiratory Mechanics , Time Factors
13.
Cell Stem Cell ; 22(1): 91-103.e5, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29276141

ABSTRACT

Direct cardiac reprogramming holds great promise for regenerative medicine. We previously generated directly reprogrammed induced cardiomyocyte-like cells (iCMs) by overexpression of Gata4, Mef2c, and Tbx5 (GMT) using retrovirus vectors. However, integrating vectors pose risks associated with insertional mutagenesis and disruption of gene expression and are inefficient. Here, we show that Sendai virus (SeV) vectors expressing cardiac reprogramming factors efficiently and rapidly reprogram both mouse and human fibroblasts into integration-free iCMs via robust transgene expression. SeV-GMT generated 100-fold more beating iCMs than retroviral-GMT and shortened the duration to induce beating cells from 30 to 10 days in mouse fibroblasts. In vivo lineage tracing revealed that the gene transfer of SeV-GMT was more efficient than retroviral-GMT in reprogramming resident cardiac fibroblasts into iCMs in mouse infarct hearts. Moreover, SeV-GMT improved cardiac function and reduced fibrosis after myocardial infarction. Thus, efficient, non-integrating SeV vectors may serve as a powerful system for cardiac regeneration.


Subject(s)
Cellular Reprogramming , Genetic Vectors/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Sendai virus/genetics , Action Potentials , Animals , Animals, Newborn , Cell Lineage , Cell Proliferation , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Humans , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Transcription Factors/metabolism , Transgenes , Virion/metabolism
14.
Oncol Rep ; 39(1): 173-181, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29192325

ABSTRACT

Our previous studies of the microRNA (miRNA) expression signature in clear cell renal cell carcinoma (ccRCC) indicated that miRNA-1274a (miR-1274a) was significantly upregulated in clinical specimens, suggesting that miR-1274a may act as an oncogenic miRNA in ccRCC. The aim of this study was to investigate the functional roles of miR-1274a and identify downstream tumor-suppressive targets regulated by miR­1274a in ccRCC cells. Functional studies of miR-1274a were carried out by anti-miRNA to investigate cell proliferation and apoptosis using the A498, ACHN and Caki1 ccRCC cell lines. Suppression of miR-1274a significantly inhibited cancer cell proliferation and induced apoptosis in the ccRCC cells. Gene expression data combined with in silico analysis and luciferase reporter assays demonstrated that bone morphogenetic protein receptor type 1B (BMPR1B) was directly regulated by miR-1274a. Moreover, TCGA database as well as immunohistochemistry demonstrated low expression of BMPR1B in ccRCC clinical specimens compared to that in normal kidney tissues. We conclude that loss of oncogenic miR-1274a reduced cancer cell proliferation and induced apoptosis in ccRCC through targeting BMPR1B. Our data revealing molecular pathways and a target gene regulated by oncogenic miR-1274a provide new insight into the potential mechanisms of ccRCC oncogenesis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Carcinoma, Renal Cell/genetics , Down-Regulation , Kidney Neoplasms/genetics , MicroRNAs/genetics , 3' Untranslated Regions , A549 Cells , Aged , Apoptosis , Bone Morphogenetic Protein Receptors, Type I/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Survival Analysis
15.
Cancer Res ; 77(22): 6321-6329, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28951458

ABSTRACT

Continuous activation of hypoxia-inducible factor (HIF) is important for progression of renal cell carcinoma (RCC) and acquired resistance to antiangiogenic multikinase and mTOR inhibitors. Recently, HIF2α antagonists PT2385 and PT2399 were developed and are being evaluated in a phase I clinical trial for advanced or metastatic clear cell RCC (ccRCC). However, resistance to HIF2α antagonists would be expected to develop. In this study, we identified signals activated by HIF2α deficiency as candidate mediators of resistance to the HIF2α antagonists. We established sunitinib-resistant tumor cells in vivo and created HIF2α-deficient variants of these cells using CRISPR/Cas9 technology. Mechanistic investigations revealed that a regulator of the serine biosynthesis pathway, phosphoglycerate dehydrogenase (PHGDH), was upregulated commonly in HIF2α-deficient tumor cells along with the serine biosynthesis pathway itself. Accordingly, treatment with a PHGDH inhibitor reduced the growth of HIF2α-deficient tumor cells in vivo and in vitro by inducing apoptosis. Our findings identify the serine biosynthesis pathway as a source of candidate therapeutic targets to eradicate advanced or metastatic ccRCC resistant to HIF2α antagonists. Cancer Res; 77(22); 6321-9. ©2017 AACR.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Phosphoglycerate Dehydrogenase/metabolism , Serine/biosynthesis , Xenograft Model Antitumor Assays/methods , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CRISPR-Cas Systems , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indans/pharmacology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy/methods , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Phosphoglycerate Dehydrogenase/genetics , Sulfones/pharmacology , Survival Analysis
16.
Br J Cancer ; 116(8): 1077-1087, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28324890

ABSTRACT

BACKGROUND: Based on the microRNA (miRNA) signature of bladder cancer (BC) by deep sequencing, we recently found that several double-stranded mature miRNAs derived from the same pre-miRNAs were sufficiently expressed and acted as tumour suppressors by regulating common target genes in BC. Our deep-sequencing signature of BC showed that all miR-199 family members (miR-199a-3p/-5p and miR-199b-3p/-5p) were also downregulated. We hypothesised that these miRNAs may function as tumour suppressors by regulating common target genes. METHODS: Functional assays of BC cells were performed using transfection of mature miRNA. In silico analyses and luciferase reporter analyses were applied to identify target genes of these miRNAs. The overall survival of patients with BC in The Cancer Genome Atlas (TCGA) database was evaluated by the Kaplan-Meier method. RESULTS: Restoration of these miRNAs significantly inhibited cell migration and invasion in BC cells. Integrin α3 (ITGA3) was directly regulated by these miRNAs. The Cancer Genome Atlas database showed that patients with low pre-miR-199 family (miR-199a-1/-2 and miR-199b) expression exhibited significantly poorer overall survival compared with patients with high pre-miR-199 family expression. CONCLUSIONS: miR-199 family miRNAs functioned as tumour suppressors in BC cells by targeting ITGA3 and might be good prognostic markers for predicting survival in patients with BC.


Subject(s)
Gene Expression Regulation, Neoplastic , Integrin alpha3/metabolism , MicroRNAs/genetics , Urinary Bladder Neoplasms/genetics , Apoptosis , Biomarkers, Tumor , Blotting, Western , Cell Movement , Cell Proliferation , Humans , Immunoenzyme Techniques , Integrin alpha3/genetics , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
17.
Oncotarget ; 8(13): 20881-20894, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28152509

ABSTRACT

Previous studies showed that five miRNAs (miR-885-5p, miR-1274, miR-210-3p, miR-224 and miR-1290) were upregulated the most in clear cell renal cell carcinoma (ccRCC). Our focus was to understand from a clinical standpoint the functional consequences of upregulating miR-210-3p. Towards this, we utilized the CRISPR/Cas9 gene editing system to deplete miR-210-3p in RCC cell lines (786-o, A498 and Caki2) and characterized the outcomes. We observed that miR-210-3p depletion dramatically increased tumorigenesis, including altering the morphology of A498 and Caki2 cells in a manner characteristic of epithelial-mesenchymal transition (EMT). These results were corroborated by in vivo xenograft studies, which showed enhanced growth of tumors from miR-210-3p-depleted A498 cells. We identified Twist-related protein 1 (TWIST1) as a key target of miR-210-3p. Analysis of the ccRCC patient data in The Cancer Genome Atlas database showed a negative correlation between miR-210-3p and TWIST1 expression. High TWIST1 and low miR-210-3p expression associated with poorer overall and disease-free survival as compared to low TWIST1 and high miR-210-3p expression. These findings suggest that renal cell carcinoma progression is promoted by TWIST1 suppression mediated by miR-210-3p.


Subject(s)
CRISPR-Cas Systems/genetics , Carcinoma, Renal Cell/pathology , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , MicroRNAs/antagonists & inhibitors , Nuclear Proteins/metabolism , Twist-Related Protein 1/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition , Female , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Staging , Nuclear Proteins/genetics , Prognosis , Survival Rate , Tumor Cells, Cultured , Twist-Related Protein 1/genetics , Xenograft Model Antitumor Assays
18.
J Endourol ; 31(4): 391-395, 2017 04.
Article in English | MEDLINE | ID: mdl-28049342

ABSTRACT

PURPOSE: When accidental tumor incision (ATI) has occurred during open partial nephrectomy (PN), scissors can be changed easily. In contrast, during laparoscopic partial nephrectomy (LPN) or robotic partial nephrectomy (RPN), it is time consuming and expensive especially during RPN to change scissors. This study investigates whether tumor cells remain on the surface of scissors after ATI during PN and investigates an alternative way to avoid changing scissors during LPN and RPN. MATERIALS AND METHODS: We subcutaneously injected 786-O renal-cell carcinoma (RCC) cells containing enhanced green fluorescent protein (786-O/EGFP) into six mice. We incised the subsequent tumor with straight or Microline scissors. The scissor surfaces were then examined by microscopy for detection of EGFP immunofluorescence. In addition, the scissor surfaces were treated in three ways: no electrical treatment, electrical treatment of 20 W for 5 seconds, and electrical treatment of 40 W for 5 seconds. RESULTS: Strings or dots of EGFP were detected on every scissor surface, and 786-O/EGFP cells were alive and able to proliferate in medium in 33% of the nonelectrically treated samples. However, no 786-O/EGFP cells treated with monopolar electricity survived. In another experiment, we also found that 100 786-O cells placed on scissor surfaces could not survive after the same electrical treatment. CONCLUSIONS: RCC cells remained on scissors after ATI; however, electrical treatment eliminated tumor cells, possibly preventing recurrence or metastasis after surgery.


Subject(s)
Carcinoma, Renal Cell/surgery , Equipment Contamination , Kidney Neoplasms/surgery , Nephrectomy/methods , Robotic Surgical Procedures/methods , Surgical Instruments , Animals , Cell Line, Tumor , Electricity , Female , Humans , In Vitro Techniques , Laparoscopy/methods , Mice , Mice, Nude , Neoplasm Recurrence, Local , Neoplasm Transplantation , Retrospective Studies , Robotics
19.
Cancer Sci ; 107(9): 1233-42, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27355528

ABSTRACT

Our recent study of the microRNA (miRNA) expression signature of bladder cancer (BC) by deep-sequencing revealed that two miRNA, microRNA-139-5p/microRNA-139-3p were significantly downregulated in BC tissues. The aim of this study was to investigate the functional roles of these miRNA and their modulation of cancer networks in BC cells. Functional assays of BC cells were performed using transfection of mature miRNA or small interfering RNA (siRNA). Genome-wide gene expression analysis, in silico analysis and dual-luciferase reporter assays were applied to identify miRNA targets. The associations between the expression of miRNA and its targets and overall survival were estimated by the Kaplan-Meier method. Gain-of-function studies showed that miR-139-5p and miR-139-3p significantly inhibited cell migration and invasion by BC cells. The matrix metalloprotease 11 gene (MMP11) was identified as a direct target of miR-139-5p and miR-139-3p. Kaplan-Meier survival curves showed that higher expression of MMP11 predicted shorter survival of BC patients (P = 0.029). Downregulated miR-139-5p or miR-139-3p enhanced BC cell migration and invasion in BC cells. MMP11 was directly regulated by these miRNA and might be a good prognostic marker for survival of BC patients.


Subject(s)
Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 11/genetics , MicroRNAs/genetics , RNA Interference , Urinary Bladder Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Female , Gene Expression , Gene Expression Profiling , Gene Knockdown Techniques , Genes, Reporter , Humans , Immunohistochemistry , Male , Prognosis , RNA, Messenger/genetics , Transfection , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
20.
Int J Oncol ; 49(1): 111-22, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27212625

ABSTRACT

Advanced prostate cancer (PCa) metastasizes to bone and lymph nodes, and currently available treatments cannot prevent the progression and metastasis of the disease. Therefore, an improved understanding of the molecular mechanisms of the progression and metastasis of advanced PCa using current genomic approaches is needed. Our miRNA expression signature in castration-resistant prostate cancer (CRPC) revealed that microRNA-320a (miR­320a) was significantly reduced in cancer tissues, suggesting that miR­320a may be a promising anticancer miRNA. The aim of this study was to investigate the functional roles of miR­320a in naïve PCa and CRPC cells and to identify miR­320a-regulated genes involved in PCa metastasis. The expression levels of miR­320a were significantly reduced in naïve PCa, CRPC specimens, and PCa cell lines. Restoration of mature miR­320a in PCa cell lines showed that miR­320a significantly inhibited cancer cell migration and invasion. Moreover, we found that lysosomal-associated membrane protein 1 (LAMP1) was a direct target of miR­320a in PCa cells. Silencing of LAMP1 using siRNA significantly inhibited cell proliferation, migration, and invasion in PCa cells. Overexpression of LAMP1 was observed in PCa and CRPC clinical specimens. Moreover, downstream pathways were identified using si-LAMP1-transfected cells. The discovery of tumor-suppressive miR­320a-mediated pathways may provide important insights into the potential mechanisms of PCa metastasis.


Subject(s)
Cell Proliferation/genetics , Lysosomal Membrane Proteins/genetics , MicroRNAs/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Humans , Lysosomal Membrane Proteins/biosynthesis , Male , MicroRNAs/biosynthesis , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Prostatic Neoplasms, Castration-Resistant/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...