Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 28(7): e18235, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509735

ABSTRACT

Kidney stone, one of the oldest known diseases, has plagued humans for centuries, consistently imposing a heavy burden on patients and healthcare systems worldwide due to their high incidence and recurrence rates. Advancements in endoscopy, imaging, genetics, molecular biology and bioinformatics have led to a deeper and more comprehensive understanding of the mechanism behind nephrolithiasis. Kidney stone formation is a complex, multi-step and long-term process involving the transformation of stone-forming salts from free ions into asymptomatic or symptomatic stones influenced by physical, chemical and biological factors. Among the various types of kidney stones observed in clinical practice, calcareous nephrolithiasis is currently the most common and exhibits the most intricate formation mechanism. Extensive research suggests that calcareous nephrolithiasis primarily originates from interstitial subepithelial calcified plaques and/or calcified blockages in the openings of collecting ducts. These calcified plaques and blockages eventually come into contact with urine in the renal pelvis, serving as a nidus for crystal formation and subsequent stone growth. Both pathways of stone formation share similar mechanisms, such as the drive of abnormal urine composition, involvement of oxidative stress and inflammation, and an imbalance of stone inhibitors and promoters. However, they also possess unique characteristics. Hence, this review aims to provide detailed description and present recent discoveries regarding the formation processes of calcareous nephrolithiasis from two distinct birthplaces: renal interstitium and tubule lumen.


Subject(s)
Calcinosis , Kidney Calculi , Humans , Kidney Medulla/metabolism , Kidney Calculi/complications , Kidney Calculi/metabolism , Calcinosis/metabolism , Endoscopy , Inflammation/metabolism
2.
Adv Sci (Weinh) ; 11(17): e2309234, 2024 May.
Article in English | MEDLINE | ID: mdl-38380498

ABSTRACT

The CRISPR-Cas system, initially for DNA-level gene editing and transcription regulation, has expanded to RNA targeting with the Cas13d family, notably the RfxCas13d. This advancement allows for mRNA targeting with high specificity, particularly after catalytic inactivation, broadening the exploration of translation regulation. This study introduces a CRISPR-dCas13d-eIF4G fusion module, combining dCas13d with the eIF4G translation regulatory element, enhancing target mRNA translation levels. This module, using specially designed sgRNAs, selectively boosts protein translation in targeted tissue cells without altering transcription, leading to notable protein expression upregulation. This system is applied to a kidney stone disease model, focusing on ferroptosis-linked GPX4 gene regulation. By targeting GPX4 with sgRNAs, its protein expression is upregulated in human renal cells and mouse kidney tissue, countering ferroptosis and resisting calcium oxalate-induced cell damage, hence mitigating stone formation. This study evidences the CRISPR-dCas13d-eIF4G system's efficacy in eukaryotic cells, presenting a novel protein translation research approach and potential kidney stone disease treatment advancements.


Subject(s)
CRISPR-Cas Systems , Calcium Oxalate , Disease Models, Animal , Eukaryotic Initiation Factor-4G , Ferroptosis , Ferroptosis/genetics , Mice , Animals , Calcium Oxalate/metabolism , CRISPR-Cas Systems/genetics , Humans , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Kidney Calculi/genetics , Kidney Calculi/metabolism , Protein Biosynthesis/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
3.
J Cell Mol Med ; 28(5): e18101, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38165009

ABSTRACT

Bladder cancer is one of the most common malignant tumours of the urogenital system, with high morbidity and mortality. In most cases, surgery is considered the first choice of treatment, followed by adjuvant chemotherapy. However, the 5-year recurrence rate is still as high as 65% in patients with non-invasive or in situ tumours and up to 73% in patients with slightly more advanced disease at initial diagnosis. Various treatment methods for bladder cancer have been developed, and hundreds of new immunotherapies are being tested. To date, only a small percentage of people have had success with new treatments, though studies have suggested that the combination of immunotherapy with other therapies improves treatment efficiency and positive outcomes for individuals, with great hopes for the future. In this article, we summarize the origins, therapeutic mechanisms and current status of research on immunotherapeutic agents for bladder cancer.

5.
Urolithiasis ; 51(1): 118, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37796347

ABSTRACT

The Holmium (Ho:YAG) laser is presently the most extensively employed in laser lithotripsy for the management of kidney stones. Despite its adoption as the gold standard for laser lithotripsy, Ho:YAG laser lithotripsy poses three significant challenges, namely thermal effect, insufficient stone fragmentation, and stone displacement, which have garnered increased attention from urologic surgeons. Nowadays, the femtosecond laser is regarded as a potential alternative to the Ho:YAG laser due to its capacity to ablate diverse materials with minimal thermal effect. In our ex vivo investigation, we assessed the dimensions of ablation pits, the efficacy of ablation, the degree of stone fragmentation, the alterations in water temperature surrounding stones, and the degree of tissue damage associated with Femtosecond laser lithotripsy utilizing adjustable power settings (1-50 W). Our findings indicate that the ablation pits generated by the Femtosecond laser exhibited uniform geometries, and the effectiveness of ablation and fragmentation for Femtosecond laser lithotripsy were significantly and positively correlated with laser power. When the laser power remained constant, the Femtosecond laser with higher pulse energy demonstrated superior efficiency in stone ablation, but inferior performance in stone fragmentation. Conversely, the Femtosecond laser with higher pulse frequency exhibited the opposite behavior. Furthermore, the thermal effect increased proportionally with laser power, leading to a tentative recommendation of 10W laser power for future investigations. Our in vitro findings suggest that the Femtosecond laser holds promise as a safe and effective alternative to holmium lasers.


Subject(s)
Kidney Calculi , Lasers, Solid-State , Lithotripsy, Laser , Lithotripsy , Humans , Lithotripsy, Laser/adverse effects , Lithotripsy, Laser/methods , Lithotripsy/adverse effects , Kidney Calculi/surgery , Lasers, Solid-State/therapeutic use , Holmium
6.
Cell Mol Life Sci ; 80(10): 301, 2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37740796

ABSTRACT

Surgical crushing of stones alone has not addressed the increasing prevalence of kidney stones. A promising strategy is to tackle the kidney damage and crystal aggregation inherent in kidney stones with the appropriate therapeutic target. FKBP prolyl isomerase 5 (FKBP5) is a potential predictor of kidney injury, but its status in calcium oxalate (CaOx) kidney stones is not clear. This study attempted to elucidate the role and mechanism of FKBP5 in CaOx kidney stones. Lentivirus and adeno-associated virus were used to control FKBP5 expression in a CaOx kidney stone model. Transcriptomic sequencing and immunological assays were used to analyze the mechanism of FKBP5 deficiency in CaOx kidney stones. The results showed that FKBP5 deficiency reduced renal tubular epithelial cells (RTEC) apoptosis and promoted cell proliferation by downregulating BOK expression. It also attenuated cell-crystal adhesion by downregulating the expression of CDH4. In addition, it inhibited M1 polarization and chemotaxis of macrophages by suppressing CXCL10 expression in RTEC. Moreover, the above therapeutic effects were exerted by inhibiting the activation of NF-κB signaling. Finally, in vivo experiments showed that FKBP5 deficiency attenuated stone aggregation and kidney injury in mice. In conclusion, this study reveals that FKBP5 deficiency attenuates cell-crystal adhesion, reduces apoptosis, promotes cell proliferation, and inhibits macrophage M1 polarization and chemotaxis by inhibiting NF-κB signaling. This provides a potential therapeutic target for CaOx kidney stones.


Subject(s)
Kidney Calculi , NF-kappa B , Animals , Mice , Calcium Oxalate , Signal Transduction , Kidney Calculi/genetics , Apoptosis
7.
World J Surg Oncol ; 21(1): 271, 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37641150

ABSTRACT

OBJECTIVE: Radical cystectomy (RC) is the gold standard treatment for muscle-invasive bladder cancer (MIBC). As a bladder-preservation option recommended in guidelines, trimodal therapy (TMT) has become increasingly popular in recent years. However, it is still uncertain whether TMT can provide comparable oncologic outcomes to RC. Therefore, it is imperative to evaluate whether TMT yields comparable outcomes to RC. METHODS: We conducted a systematic search of Web of Science, MEDLINE, the Cochrane Library, and EMBASE databases up to June 2023 to identify studies that met our inclusion criteria. The primary outcome measures evaluated in this study were overall survival (OS) and cancer-specific survival (CSS). The study quality was evaluated independently by two authors, and data were extracted accordingly. RESULTS: After excluding duplicates and ineligible articles, our meta-analysis included seven studies involving 3,489 and 13,877 patients in the TMT and RC groups, respectively. Short-term overall survival rates were comparable between the groups, but beyond 5 and > 10-years, the RC group had significantly higher overall survival rates compared to the TMT group. In terms of cancer-specific survival, there was no significant difference between the groups at 1-year follow-up, but from the second year onwards, including the 5-year and > 10-year nodes, the RC group had significantly better outcomes compared to the TMT group. CONCLUSION: The treatment effect of RC is better than that of TMT. Unless the patient can't tolerate RC or has a strong desire to preserve the bladder, RC should be chosen over TMT in treatment, and patients undergoing TMT should be closely followed up.


Subject(s)
Urinary Bladder Neoplasms , Urinary Bladder , Humans , Cystectomy , Databases, Factual , Muscles , Urinary Bladder Neoplasms/therapy
8.
Int Immunopharmacol ; 124(Pt A): 110801, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37651854

ABSTRACT

Hyperoxaluria-induced damage to renal tubular epithelial cells (RTECs) is considered the most significant contributor to kidney stone formation. However, the precise regulatory mechanism underlying this damage, particularly its association with mitophagy dysfunction, remains unclear. Additionally, effective preventive medications for kidney stones are lacking. Melatonin, a hormone secreted by the pituitary gland that primarily regulates circadian rhythm, has been found to modulate mitophagy in recent research. Therefore, this investigation aims to examine the impact of melatonin on mitophagy and cellular impairment in the formation of kidney stone. The results of this study reveal that melatonin can alleviate the formation of kidney stones and reduce oxalate-induced renal injuries. In the RTECs of kidney stone model, mitophagy was found to be impaired, leading to increased oxidative stress, inflammation, and ferroptosis both in vivo and in vitro. Melatonin was shown to have a restorative potential in enhancing PINK1-Parkin-regulated mitophagy through AMPK phosphorylation, reducing excessive ROS release and inhibiting oxidative stress, inflammation and ferroptosis. Further experiments demonstrated that the protective effect of melatonin was diminished by PINK1 knockdown and AMPK pathway blockade. This study is the first to reveal the interplay between mitophagy and ferroptosis in kidney stone models and establish the protective role of melatonin in restoring mitophagy to inhibit ferroptosis.

9.
Front Cell Infect Microbiol ; 13: 1142426, 2023.
Article in English | MEDLINE | ID: mdl-37265501

ABSTRACT

Background: The identification of uropathogens (UPBs) and urinary tract colonizing bacteria (UCB) conduces to guide the antimicrobial therapy to reduce resistant bacterial strains and study urinary microbiota. This study established a nomogram based on the nanopore-targeted sequencing (NTS) and other infectious risk factors to distinguish UPB from UCB. Methods: Basic information, medical history, and multiple urine test results were continuously collected and analyzed by least absolute shrinkage and selection operator (LASSO) regression, and multivariate logistic regression was used to determine the independent predictors and construct nomogram. Receiver operating characteristics, area under the curve, decision curve analysis, and calibration curves were used to evaluate the performance of the nomogram. Results: In this study, the UPB detected by NTS accounted for 74.1% (401/541) of all urinary tract microorganisms. The distribution of ln(reads) between UPB and UCB groups showed significant difference (OR = 1.39; 95% CI, 1.246-1.551, p < 0.001); the reads number in NTS reports could be used for the preliminary determination of UPB (AUC=0.668) with corresponding cutoff values being 7.042. Regression analysis was performed to determine independent predictors and construct a nomogram, with variables ranked by importance as ln(reads) and the number of microbial species in the urinary tract of NTS, urine culture, age, urological neoplasms, nitrite, and glycosuria. The calibration curve showed an agreement between the predicted and observed probabilities of the nomogram. The decision curve analysis represented that the nomogram would benefit clinical interventions. The performance of nomogram with ln(reads) (AUC = 0.767; 95% CI, 0.726-0.807) was significantly better (Z = 2.304, p-value = 0.021) than that without ln(reads) (AUC = 0.727; 95% CI, 0.681-0.772). The rate of UPB identification of nomogram was significantly higher than that of ln(reads) only (χ2 = 7.36, p-value = 0.009). Conclusions: NTS is conducive to distinguish uropathogens from colonizing bacteria, and the nomogram based on NTS and multiple independent predictors has better prediction performance of uropathogens.


Subject(s)
Microbiota , Nanopore Sequencing , Nanopores , Nomograms , Bacteria/genetics
11.
Urolithiasis ; 51(1): 71, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37067622

ABSTRACT

The high incidence, recurrence and treatment costs of urolithiasis have a serious impact on patients and society. For a long time, countless scholars have been working tirelessly on studies related to the etiology of urolithiasis. A comprehensive understanding of the current status will be beneficial to the development of this field. We collected all literature about the etiology of urolithiasis from 1990 to 2022 using the Web of Science (WoS) database. VOSviewer, Bibliometrix and CiteSpace software were used to quantitatively analyze and visualize the data as well. The query identified 3177 articles for final analysis, of which related to the etiology of urolithiasis. The annual number of publications related to urolithiasis research has steadily increased during the latest decade. United States (1106) and China (449) contributed the most publications. University of Chicago (92) and Indiana University (86) have the highest number of publications. Urolithiasis and Journal of Urology have published the most articles in the field. Coe FL is the most productive author (63 articles), whose articles have obtained the most citations in all (4141 times). The keyword, such as hypercalciuria, hyperoxaluria, citrate, oxidative stress, inflammation, Randall's plaque, are the most attractive targets for the researchers. Our review provides a global landscape of studies related to the etiology of urolithiasis, which can serve as a reference for future studies in this field.


Subject(s)
Bibliometrics , Urolithiasis , Humans , China , Databases, Factual , Urolithiasis/etiology
12.
Int Immunopharmacol ; 117: 110042, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36940552

ABSTRACT

Schizandrin B (SchB) protects against oxidative, inflammatory, and ferroptotic injury. Oxidative stress and inflammation are indispensably involved in nephrolithiasis and ferroptosis also plays an important role in stone formation. It is unclear whether SchB can ameliorate nephrolithiasis; its underlying mechanism is also unknown. First, we employed bioinformatics to investigate the mechanisms of nephrolithiasis. To evaluate the efficacy of SchB, HK-2 cell models of oxalate-induced damage, Erastin-induced ferroptosis, and the Sprague Dawley rat model of Ethylene Glycol-induced nephrolithiasis were established. Then, Nrf2 siRNA and GSK3ß overexpression plasmids were transfected into HK-2 cells to elucidate the role of SchB in regulating oxidative stress-mediated ferroptosis. In our study, oxidative stress and inflammation were strongly associated with nephrolithiasis. Administration of SchB attenuated the cell viability, dysfunctional mitochondria, oxidative stress and inflammatory response in vitro and alleviated renal injury and crystal deposition in vivo. SchB treatment also reduced the levels of cellular Fe2+ accumulation, lipid peroxidation and MDA, and regulated ferroptosis-related proteins, including XCT, GPX4, FTH1 and CD71, in Erastin-induced or oxalate-induced HK-2 cells. Mechanistically, SchB facilitated Nrf2 nuclear translocation, and silencing Nrf2 or overexpressing GSK3ß worsened oxalate-induced oxidative injury and abolished the beneficial effect of SchB against ferroptosis in vitro. To summarize, SchB could alleviate nephrolithiasis by positively regulating GSK3ß/Nrf2 signaling-mediated ferroptosis.


Subject(s)
Ferroptosis , Nephrolithiasis , Rats , Animals , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Glycogen Synthase Kinase 3 beta , Rats, Sprague-Dawley , Inflammation , Oxalates/pharmacology
13.
Arch Biochem Biophys ; 727: 109325, 2022 09 30.
Article in English | MEDLINE | ID: mdl-35749806

ABSTRACT

BACKGROUND: The ossification of renal tubular epithelial cells (RTECs) plays an important initial role in the formation of kidney stones, but its specific mechanism is still unclear. The JAK2/STAT3 signaling pathway is important for bone cell differentiation. Accordingly, we explored the role and mechanism of the JAK2/STAT3 signaling pathway in the ossification of RTECs. METHODS: We used oxalate or ethylene glycol to construct kidney stone models in vitro and in vivo, and investigated the expression of osteogenic-specific genes, osteogenesis ability, and JAK2/STAT3 signaling in the kidney stone models by western blotting, qRT-PCR, immunofluorescence, and immunohistochemistry. Then, genetic engineering or drugs were used to inhibit the expression or activation of JAK2, and the expression of osteogenic-specific genes and the osteogenic ability of the RTECs were determined again. RESULTS: In the in vitro and in vivo kidney stone models, the expression of osteogenic specific genes in the RTECs was significantly upregulated, the osteogenic capacity was significantly increased, and the expression of p-JAK2 (phospho-JAK2) and p-STAT3 (phospho-STAT3) was significantly increased. When the expression or activation of JAK2 was inhibited, the ossification of RTECs and the formation of kidney stones was reversed. CONCLUSIONS: During the formation of kidney stones, RTECs undergo obvious ossification, and the JAK2/STAT3 signaling pathway plays a key positive regulatory role in this process.


Subject(s)
Kidney Calculi , Osteogenesis , Cell Differentiation , Humans , Janus Kinase 2/metabolism , Oxalates , STAT3 Transcription Factor/metabolism , Signal Transduction
14.
Oxid Med Cell Longev ; 2021: 6630343, 2021.
Article in English | MEDLINE | ID: mdl-34659638

ABSTRACT

Renal tubular epithelial cell damage is the basis for the formation of kidney stones. Oxalate can induce human proximal tubular (HK-2) cells to undergo autophagy and ferroptosis. The present study was aimed at investigating whether the ferroptosis of HK-2 cells induced by oxalate is caused by the excessive activation of autophagy. We treated HK-2 cells with 2 mmol/L of oxalate to establish a kidney stone model. First, we tested the degree of oxidative damage and the level of autophagy and ferroptosis in the control group and the oxalate intervention group. We then knocked down and overexpressed the BECN1 gene and knocked down the NCOA4 gene in HK-2 cells, followed by redetection of the above indicators. We confirmed that oxalate could induce autophagy and ferroptosis in HK-2 cells. Moreover, after oxalate treatment, overexpression of the BENC1 gene increased cell oxidative damage and ferroptosis. In addition, knockdown of NCOA4 reversed the effect of oxalate-induced ferroptosis in HK-2 cells. Our results show that the effects of oxalate on the ferroptosis of HK-2 cells are caused by the activation of autophagy, and knockdown of the NCOA4 could ameliorate this effect.


Subject(s)
Epithelial Cells/metabolism , Ferroptosis/physiology , Kidney Calculi/physiopathology , Oxalates/chemistry , Animals , Autophagy , Humans , Male , Rats , Rats, Sprague-Dawley , Transfection
15.
Int J Mol Med ; 47(1): 289-301, 2021 01.
Article in English | MEDLINE | ID: mdl-33416117

ABSTRACT

Ferroptosis is an iron­dependent lipid peroxidation process. Although the involvement of ferroptosis in kidney diseases has recently been reported, the association between ferroptosis and urolithiasis remains unclear. The present study examined the effects of ferroptosis on calcium oxalate (CaOx) crystal­induced renal tubular epithelial cell injury in vivo and in vitro. First, renal tubular epithelial cells were exposed to various concentrations of CaOx. By measuring cell viability, Fe2+ levels, lipid peroxidation levels and the levels of ferroptosis­related proteins, it was identified that the relative expression of the ferroptosis agonist proteins, p53, long­chain acyl­CoA synthetases (ACSL4), transferrin (TF) and transferrin receptor (TRC), increased, while the relative expression of the ferroptosis inhibitory proteins, solute carrier family 7 member 11 (SLC7A11, XCT) and glutathione peroxidase 4 (GPX4), decreased significantly. Furthermore, the levels of Fe2+ and lipid peroxidation gradually increased, while cell viability significantly decreased. From these results, it was noted that the extent of CaOx­induced ferroptosis activation and cell injury was dependent on the CaOx concentration. To further investigate the association between ferroptosis and renal tubular epithelial cell injury, the ferroptosis agonist, erastin, and the ferroptosis inhibitor, ferrostatin­1, were used to regulate the degree of ferroptosis at the same CaOx concentration in in vivo and in vitro experiments. CaOx­induced ferroptosis and damage to renal tubular epithelial cells and renal tissue were investigated. Finally, it was identified that through the regulation of ferroptosis levels, renal tubular epithelial cell injury increased significantly when the ferroptosis level increased, and vice versa. On the whole, the present results indicated that ferroptosis is essential for renal tubular epithelial cell injury induced by CaOx crystals. This finding is highly significant and promotes the further investigation of the association between ferroptosis and urolithiasis.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Calcium Oxalate/metabolism , Ferroptosis , Gene Expression Regulation , Kidney Tubules/metabolism , Urolithiasis/metabolism , Animals , Cell Line , Humans , Kidney Tubules/pathology , Male , Rats , Rats, Sprague-Dawley , Urolithiasis/pathology
16.
Cell Cycle ; 19(20): 2600-2610, 2020 10.
Article in English | MEDLINE | ID: mdl-32871086

ABSTRACT

Background: Deposition of various crystal and organic substances in the kidney can lead to kidney stone formation. Melatonin is an effective endogenous antioxidant that can prevent crystalluria and kidney damage due to crystal formation and aggregation. In this study, we investigated the mechanism by which melatonin inhibits endoplasmic reticulum (ER) stress and apoptosis. Methods: We treated HK-2 cells with oxalate to establish an in vitro kidney stone model, and treated these cells with different concentrations of melatonin (0, 5, 10, 20 µmol/L) and the AMP-activated protein kinase (AMPK) inhibitor Compound C. We measured levels of stress response markers including reactive oxygen species (ROS), lactate dehydrogenase (LDH), glutathione (GSH), superoxide dismutase (SOD), malondialdehyde (MDA), catalase (CAT), and factors in the stress response pathway, such as ATF6, GRP78, DDIT3, PERK, p-PERK, IRE1, p-IRE1, XBP1s, AMPK, and p-AMPK, using real time-PCR, western blot, and immunofluorescence analyzes. We measured mitochondrial membrane potential and caspases-3 activity using the CCK8, enzyme-linked immunosorbent, and flow cytometry assays to assess HK-2 cell viability and apoptosis. Results: Melatonin improved the total antioxidant capacity (T-AOC) of the HK-2 cells, as evidenced by the dose-dependent reduction in apoptosis, ROS levels, and protein expression of ATF6, GRP78, DDIT3, p-PERK, p-IRE1, XBP1s, caspase-12, cleaved caspase-3 and cleaved caspase-9. Addition of the AMPK inhibitor, Compound C, partially reversed the protective effect of melatonin. Conclusion: Our study revealed that the protective effects of melatonin on oxalate-induced ER stress and apoptosis is partly dependent on AMPK activation in HK-2 cells. These findings provide insight into the prevention and treatment of kidney stones.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , Melatonin/pharmacology , Oxalates/pharmacology , Signal Transduction/drug effects , Apoptosis/drug effects , Biomarkers/metabolism , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism
17.
Life Sci ; 254: 117672, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32304760

ABSTRACT

AIM: MicroRNAs (miRNAs) function as oncogenes or tumor suppressors in an extensive variety of human cancers, including prostate cancer (PCa). Herein, the aim of this study is to investigate the effect of miR-381 on autophagy and apoptosis of PCa cells through the reelin (RELN)-mediated the PI3K/AKT/mTOR signaling pathway. METHODS: PCa-related differentially expressed genes and regulatory miRNA were retrieved according to microarray-based analysis and online mRNA-miRNA interaction analysis. The regulatory relationship between miR-381 and RELN was verified by bioinformatics prediction and dual-luciferase reporter gene assay. Gain- and loss-of-function experiments were applied to analyze the effects of miR-381 and RELN on the abilities of proliferation, apoptosis as well as autophagy of PCa cells. Furthermore, xenograft tumor was developed in nude mice to examine the tumorigenic ability of PCa. KEY FINDINGS: RELN, which was found to be up-regulated in PCa, was identified as a target of miR-381. Overexpressed miR-381 suppressed PCa cell proliferation while promoted autophagy and apoptosis of PCa cells. In addition, miR-381 inhibited the activation of the PI3K/AKT/mTOR signaling pathway by targeting RELN. In vivo experiments also confirmed that overexpression of miR-381 could inhibit the tumorigenic ability of PCa. CONCLUSION: Up-regulation of miR-381 inhibits the expression of RELN, which in turn inhibits the activation of the PI3K/AKT/mTOR signaling pathway, thereby inhibiting PCa cell proliferation, and promoting PCa cell apoptosis and autophagy.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , MicroRNAs/physiology , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Humans , Male , Reelin Protein
18.
Biomed Res Int ; 2019: 6304851, 2019.
Article in English | MEDLINE | ID: mdl-31275981

ABSTRACT

BACKGROUND: Inducible nitric oxide synthase (iNOS) is confirmed to regulate the production of nitric oxide (NO) when cells are exposed to external stimulus. Recent publications revealed that overexpression of iNOS predicted poor clinical outcomes for patients with malignant cancers, e.g., gastric, bladder, and colorectal cancers; however, several studies reported no obvious relationship between iNOS expression and prognosis of solid tumors. The aim of our study was to investigate the pooled effect of the prognostic value of iNOS expression. MATERIALS AND METHODS: We performed a systematic search of PubMed, Web of Science, and Embase databases up to January 15, 2019. The concerned outcomes of interest included overall survival (OS), cancer-special survival (CSS), and recurrence-free survival (RFS). RESULTS: Fourteen studies with 1,758 patients were included in this meta-analysis, and we reached the conclusion that increased iNOS expression was significantly associated with worse OS (HR: 1.89, 95% CI: 1.57 - 2.28, p ≤ 0.001), worse CSS (HR: 3.13, 95% CI: 1.88 - 5.20, p ≤ 0.001), and worse RFS (HR: 2.16, 95% CI: 1.29 - 3.62, p = 0.003) in solid tumors. Furthermore, the subgroup analysis identified the significant relationship of high iNOS expression with poor OS in gastric cancer. No obvious publication bias was detected by Begg's tests. CONCLUSION: In summary, the results drawn in our meta-analysis demonstrated that elevated expression of iNOS had a significant association with poor survival in human cancer. iNOS might serve as a promising predictive biomarker of prognosis in cancer patients, and well-designed prospective studies are further needed to substantiate the prognostic value of iNOS.


Subject(s)
Neoplasms/diagnosis , Neoplasms/enzymology , Nitric Oxide Synthase Type II/metabolism , Disease-Free Survival , Humans , Prognosis , Publication Bias
19.
J Cell Physiol ; 234(8): 12910-12917, 2019 08.
Article in English | MEDLINE | ID: mdl-30536843

ABSTRACT

Long noncoding RNA (lncRNA) exerts an essential role in the pathological processes of many diseases. Our previous study found that lncRNA ATB was highly expressed in renal cell carcinoma (RCC). Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), and migration-related assays were conducted to access the regulatory effects of lncRNA ATB on proliferative and migratory capacities of RCC cells. Flow cytometry was carried out to determine cell cycle and apoptosis influenced by lncRNA ATB. The interaction among lncRNA ATB, DNMT1, and p53 was evaluated through RNA immunoprecipitation (RIP), chromatin immunoprecipitation (ChIP), and western blot analyses. The results showed that lncRNA ATB knockdown in RCC cell line ACHN inhibited proliferative and migratory capacities and promoted apoptosis. Meanwhile, overexpression of lncRNA ATB in RCC cell line A-498 promoted proliferative and migratory capacities but inhibited apoptosis. RIP and ChIP assays confirmed that lncRNA ATB can bind to DNMT1 and stabilize its expression; meanwhile, it can promote the binding of DNMT1 to p53. Overexpression of p53 partially reversed the proliferative and migratory changes caused by lncRNA ATB. To sum up, our study revealed that high expression of lncRNA ATB could accelerate the proliferative and migratory rates of RCC cells and inhibit cell apoptosis through downregulating p53 via binding to DNMT1.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic/genetics , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis/genetics , Carcinoma, Renal Cell/pathology , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Down-Regulation/genetics , Humans
20.
Exp Biol Med (Maywood) ; 241(2): 147-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26286106

ABSTRACT

Clinically, autologous gastrointestinal segments are traditionally used for urinary diversion. However, this procedure often causes many serious complications. Tissue engineering may provide an alternative treatment method in urinary diversion. This research aims to produce tissue-engineered tubular substitutions by using homologous adipose-derived stem cells, smooth muscle cells, and bladder acellular matrix in developing urinary diversion in a rabbit model. Adipose-derived stem cells and smooth muscle cells of rabbit were obtained and cultured in vitro. These cultured adipose-derived stem cells and smooth muscle cells were seeded onto the two sides of the bladder acellular matrix and then incubated for seven days. The cell-seeded matrix was used to build tissue-engineered tubular substitutions, which were then implanted and wrapped into the omentum in vivo for two weeks to promote angiogenesis. In the experimental group, the bladder of 20 rabbits was totally resected, and the above tissue-engineered tubular substitutions were used for urinary diversion. In the control group, bladder acellular matrix tubular substitutions with unseeded cells were implanted into the omentum and were used as urinary diversion on another five rabbits with the same process. The implants were harvested, and histological examination was conducted at 2, 4, 8, and 16 weeks after operation. Intravenous urography assessment was performed at 16 weeks postoperatively. All the rabbits were alive in the experimental group until they were sacrificed. Histological analysis of the construct displayed the presence of multilayer urothelial cells on the luminal side and organized smooth muscle tissue on the other side, and different diameters of neovascularization were clearly identified in the substitutions obtained. No leakage, stricture, or obstructions were noted with intravenous urography assessment. All the animals in the control group died within two weeks, and urine leakage, scar formation, and inflammation were detected through autopsy. This study demonstrates the feasibility of tissue-engineered tubular substitutions constructed using homologous adipose-derived stem cells, smooth muscle cells, and bladder acellular matrix for urinary diversion in a rabbit model.


Subject(s)
Extracellular Matrix , Myocytes, Smooth Muscle/physiology , Stem Cells/physiology , Tissue Engineering/methods , Urinary Bladder , Urinary Diversion/methods , Animals , Male , Models, Animal , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...