Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Front Immunol ; 13: 968729, 2022.
Article in English | MEDLINE | ID: mdl-35967438

ABSTRACT

Background: Nowadays, immune checkpoint inhibitors (ICIs) have become one of the essential immunotherapies for cancer patients. However, the impact of antibiotic (ATB) use on cancer patients treated with ICIs remains controversial. Methods: Our research included retrospective studies and a randomized clinical trial (RCT) with cancer patients treated with ICIs and ATB, from the public database of PubMed, Web of Science, Embase, Cochrane, clinical trials, and JAMA. The survival outcomes included progression-free survival (PFS) and overall survival (OS). Meanwhile, hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated, and subgroup analyses were performed to determine the concrete association between ATB use and the prognosis of cancer patients treated in ICIs. Results: Our results revealed that ATB use was associated with poor survival outcomes, including OS (HR: 1.94, 95% CI: 1.68-2.25, p <0.001) and PFS (HR: 1.83, 95% CI: 1.53-2.19, p <0.001). The subgroup analysis learned about the association between ATB use and the prognosis of cancer patients with ICI treatment, including 5 cancer types, 3 kinds of ICI, 5 different ATP windows, broad-spectrum ATB class, and ECOG score. ATB treatment was associated with poor OS of non-small-cell lung cancer (NSCLC), renal cell carcinoma (RCC), esophageal cancer (EC), and melanoma (MEL) in patients treated in ICIs, while non-small-cell lung cancer (NSCLC) and renal cell carcinoma (RCC) were associated with poor PFS. Meanwhile, it was strongly related to the ICI type and ATB window. Furthermore, it is firstly mentioned that the use of broad-spectrum ATB class was strongly associated with poor PFS. Conclusion: In conclusion, our meta-analysis indicated that ATB use was significantly associated with poor OS and PFS of cancer patients treated with ICI immunotherapy, especially for patients with ATB use in the period of (-60 days; +30 days) near the initiation of ICI treatment. Also, different cancer types and the ICI type can also impact the survival outcome. This first reveals the strong relationship between the broad-spectrum ATB class and poor PFS. Still, more studies are needed for further study.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Renal Cell , Kidney Neoplasms , Lung Neoplasms , Anti-Bacterial Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Renal Cell/drug therapy , Humans , Immune Checkpoint Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Lung Neoplasms/pathology , Randomized Controlled Trials as Topic
2.
J Oncol ; 2022: 1001796, 2022.
Article in English | MEDLINE | ID: mdl-35855807

ABSTRACT

Background: To determine if the use of the Proton Pump Inhibitors (PPI) impacts the clinical efficacy of Immune Checkpoint Inhibitors (ICIs) in Non-Small Cell Lung Cancer (NSCLC), a meta-analysis was conducted. Method: Eleven studies from PubMed, EMBASE, Cochrane Library, Web of Science, and other databases up to May 2022, were selected. The pertinent clinical outcomes were assessed by applying the Progression-free survival (PFS), Overall Survival (OS), Hazard Ratio (HR), and 95% Confidence Interval (CI). Result: This study included eleven articles containing 7,893 NSCLC patients. The result indicated that PPI use was dramatically related to poor OS (HR: 1.30 [1.10-1.54]), and poor PFS (HR: 1.25 [1.09-1.42]) in case of patients treated with ICIs. With regard to the subgroup analysis, PPI use was dramatically associated with poor OS (Europe: HR = 1.48 [1.26, 1.74], Worldwide: HR = 1.54 [1.24, 1.91]), and poor PFS (Europe: HR = 1.36 [1.18, 1.57], Worldwide: HR = 1.34 [1.16, 1.55]) in patients from Europe and multi-center studies across the world, poor OS in patients with age less than or equal to 65 (HR = 1.56 [1.14, 2.15]), poor PFS in patients aged more than 65 (HR = 1.36 [1.18, 1.57]), poor OS for patients receiving with PD-1 (HR = 1.37 [1.04, 1.79]), poor PFS for patients receiving with PD-L1 (HR = 1.33 [1.19, 1.49]), and poor OS (-30: HR = 1.89 [1.29, 2.78], ±30: HR = 1.44 [1.27, 1.64]) and poor PFS (-30: HR = 1.51 [1.11, 2.05], ±30: HR = 1.32 [1.20, 1.45]) for patients who received PPI at 30 days before and/or after starting the ICIs treatment. Conclusion: Our meta-analysis indicated that PPI combined with ICIs in the treatment of NSCLC patients could result in poor OS and PFS. PPI use should be extremely cautious in clinical practices to avoid the impact on the efficacy of the ICIs.

3.
J Cosmet Dermatol ; 20(5): 1374-1380, 2021 May.
Article in English | MEDLINE | ID: mdl-33185943

ABSTRACT

BACKGROUND: Hypertrophic scar is a common complication in would healing process, and how to effectively prevent and treat it has been a hot and difficult research issue. Previous studies have showed that botulinum toxin type A (BTA) has effects on the prevention and treatment of hypertrophic scar, but little is known about the specific mechanisms. OBJECTIVE: This study aimed to explore the potential mechanisms of BTA on the inhibition of hypertrophic scar formation. METHODS: Hypertrophic scar-derived human fibroblasts were cultured and then treated with transforming growth factor-ß1 (TGF-ß1) and various concentrations of BTA. Cell proliferation and viability were measured by CellTiter 96® AQueous One Solution Cell Proliferation Assay and trypan blue staining, respectively. The total amount of collagen was examined using Sirius red staining. Collagen I and Collagen III in the culture supernatant were evaluated by enzyme-linked immunosorbent assay. Reverse transcription-quantitative polymerase chain reaction and Western blot analysis were performed to detect the transcription and translation levels. RESULTS: Our results revealed that BTA decreased the proliferation of hypertrophic scar-derived human fibroblasts. The mRNA and protein expression levels of alpha-smooth muscle actin, collagen I, and collagen III induced by TGF-ß1 were inhibited by BTA in a dose-dependent manner. BTA also inhibited the phosphorylation of Smad2/3 and ERK. CONCLUSION: BTA decreased the proliferation of fibroblasts and prevented overdeposition of ECM through the inhibition of the TGF-ß1/Smad and ERK pathways. The findings of this study provide new scientific reference for the prevention and treatment of hypertrophic scar.


Subject(s)
Botulinum Toxins, Type A , Cicatrix, Hypertrophic , Botulinum Toxins, Type A/pharmacology , Cells, Cultured , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/prevention & control , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
4.
Int J Mol Med ; 45(1): 274-275, 2020 01.
Article in English | MEDLINE | ID: mdl-31746374

ABSTRACT

Following the publication of this article, the authors noticed that the published versions of Figs. 2, 7 and 8 contained incorrect western bands. In examining their raw data, the authors realized that they had used the fibroblasts of keloids [high expression of alpha­smooth muscle actin (α­SMA)] instead of adult dermal fibroblasts (low expression of α­SMA) in certain experiments. Note that no significant differences in morphology exist between myofibroblasts (from keloids) and fibroblasts (from normal dermal tissue). These errors were brought to light since the authors identified that the expression of α­SMA in Fig. 8 was higher compared with that in Fig. 4. After careful scrutiny, they established that the first author, Bin Zhao, who performed the experiments and analyzed the data shown in Figs. 2, 7 and 8, had mislabelled the myofibroblasts as fibroblasts. However, for all the other experiments in the above­mentioned article, the cells had been used correctly. The authors regret that these errors were featured in the above­mentioned article, which may possibly have caused confusion for the readers, and the corrected versions of Figs. 2, 7 and 8 are shown opposite and on the next page. These changes did not affect either the results or the conclusions reported in this paper. The authors apologize to the Editor of International Journal of Molecular Medicine and to the readership for any inconvenience caused. [the original article was published in International Journal of Molecular Medicine 39: 153­159, 2017; DOI: 10.3892/ijmm.2016.2816].

6.
Stem Cells Int ; 2018: 5630802, 2018.
Article in English | MEDLINE | ID: mdl-30302094

ABSTRACT

Neurological diseases can severely compromise both physical and psychological health. Recently, adult mesenchymal stem cell- (MSC-) based cell transplantation has become a potential therapeutic strategy. However, most studies related to the transdifferentiation of MSCs into neural cells have had disappointing outcomes. Better understanding of the mechanisms underlying MSC transdifferentiation is necessary to make adult stem cells more applicable to treating neurological diseases. Several studies have focused on adipose-derived stromal/stem cell (ADSC) transdifferentiation. The purpose of this review is to outline the molecular characterization of ADSCs, to describe the methods for inducing ADSC transdifferentiation, and to examine factors influencing transdifferentiation, including transcription factors, epigenetics, and signaling pathways. Exploring and understanding the mechanisms are a precondition for developing and applying novel cell therapies.

7.
Cell Physiol Biochem ; 49(2): 489-500, 2018.
Article in English | MEDLINE | ID: mdl-30157481

ABSTRACT

BACKGROUND/AIMS: With increased understanding of sepsis, mortality is decreasing. However, there is still a lack of effective therapeutic strategy. The inflammatory response of macrophages is critical during sepsis. METHODS: Macrophages were stimulated with LPS. Western blotting and qRT-PCR were used to detect inflammatory responses. Then, the inhibitor of microRNA-138 was transfected and Western blotting, qRT-PCR, H&E staining and ELISA were used to verify the role of microRNA-138 in inflammation. Then target gene prediction databases were used to predict the potential target of microRNA-138. Both animal and cell models under LPS challenges were established to verify the regulation of SIRT1 and microRNA-138 during inflammation. RESULTS: The present study showed that microRNA-138 was increased in macrophages stimulated with LPS. Additionally, the NF-κB and AKT pathways were both activated. The pre-treatment of microRNA-138 inhibitor decreased inflammatory factors, downregulated the NF-κB pathway, activated the AKT pathway and protected against organ damage in mice challenged with LPS. SIRT1 was demonstrated as a potential target of microRNA-138In macrophages stimulated with LPS, the inhibition effect of microRNA-138 inhibitor on inflammation was lost by SIRT1 siRNA pre-treatment. In the animal model, the protective effect of microRNA-138 antagomir disappeared in SIRT1 knockout mice. CONCLUSION: We demonstrated that miR-138 participated in the inflammatory process by inhibiting SIRT1 and activating the NF-κB pathway.


Subject(s)
MicroRNAs/metabolism , Sirtuin 1/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/blood , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Aesthetic Plast Surg ; 42(5): 1364-1369, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30019241

ABSTRACT

BACKGROUND: Linear hypertrophic scar is a common surgical problem that can be difficult to manage, especially for the median sternotomy scar. Botulinum toxin type A (BTA) is widely used in cosmetic surgery and has been shown to improve scar quality recently. The aim of this study was to evaluate the efficacy of BTA injected in the early postoperative of median sternotomy on preventing scar formation. METHODS: In this prospective randomized controlled trial, 19 consecutive patients who underwent median sternotomy were enrolled. The median sternotomy wound in each patient was divided into the upper half and the lower half. Both halves of the wound were randomized to receive the treatment with either BTA or normal saline. At 6-month follow-up, scars were assessed using the Vancouver Scar Scale, scar widths were measured, and patients were asked to evaluate their overall satisfaction. RESULTS: Seventeen patients with median sternotomy wounds completed the entire study. At 6-month follow-up, the mean Vancouver Scar Scale score for the BTA-treated group was 3.44 ± 1.68 and for the normal saline control group was 6.29 ± 2.39, and there was a statistically significant difference between the two groups (P < 0.05). There were also significant improvements in scar width and patient satisfaction for the BTA-treated halves of the wounds (P < 0.05). CONCLUSIONS: The study demonstrates that early postoperative BTA injection can decrease scar formation and reduce scar width in median sternotomy wounds, and the overall appearance is more satisfactory. LEVEL OF EVIDENCE I: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Subject(s)
Botulinum Toxins, Type A/administration & dosage , Cicatrix/prevention & control , Sternotomy/methods , Wound Healing/drug effects , Adult , China , Double-Blind Method , Esthetics , Female , Follow-Up Studies , Humans , Injections, Intralesional , Male , Middle Aged , Prospective Studies , Reference Values , Risk Assessment , Sternotomy/adverse effects , Time Factors , Treatment Outcome
9.
Oncotarget ; 8(39): 65800-65808, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29029473

ABSTRACT

Loss of key components that form cell-cell adherens junctions, such as α-catenin, triggers severe epidermal hyperproliferation. However, the underlying molecular mechanisms remain largely unknown. We report here that neuroblastoma breakpoint family (NBPF) genes are upregulated and that NBPF7 specifically promotes cellular proliferation of α-catenin-silenced HaCaT cells through functional linkage with the NF-κB pathway. Genome-wide profiling of HaCaT cells shows that NBPF genes are upregulated following α-catenin knockdown. Data from western blot analyses are consistent with the activation of the NF-κB pathway as well as increased expression of NBPF7 by α-catenin knockdown. Co-immunoprecipitation assays indicate that NBPF7 could be detected in endogenous activated NF-κB immunoprecipitates. Immunoflurence analyses demonstrate that NBPF7 co-localizes with activated NF-κB in the nucleus after α-catenin silencing. Moreover, inhibition of NBPF7 decreases the proliferation of HaCaT cells and abolishes the enhanced proliferation associated with α-catenin knockdown in HaCaT cells. These results indicate that NBPF7 plays a key role in the α-catenin signaling pathway that regulates cell proliferation of keratinocytes. Our findings suggest that the classical NF-κB pathway plays a critical role in cellular proliferation and that NBPF7 is a functional mediator for α-catenin in the regulation of keratinocyte growth.

10.
Medicine (Baltimore) ; 96(31): e7676, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28767592

ABSTRACT

BACKGROUND: Previous studies have investigated the relationship between human bone morphogenetic protein 4 gene (BMP4) rs17563 polymorphism and nonsyndromic cleft lip with or without cleft palate (NSCL/P). However, the results remained inconsistent. Therefore, we conducted a meta-analysis to assess the effect of BMP4 rs17563 polymorphism on NSCL/P. METHODS: Electronic searches in 5 databases were conducted to select all eligible studies up to March 2017. Odds ratios (ORs) with the corresponding 95% confidence intervals (CIs) were calculated to estimate the association. Sensitivity analysis was performed to evaluate the results stability by excluding each study in turn. Publication bias was assessed by Begg funnel plots and Egger test. RESULTS: A total of 11 case-control studies were included in the meta-analysis. The pooled frequency of the minor allele C for BMP4 rs17563 was lower in Asians (pooled frequency = 0.33, 95% CI: 0.29-0.37) than in Brazilian population (pooled frequency = 0.47, 95% CI: 0.40-0.54). The overall results showed no significant association of BMP4 rs17563 polymorphism with NSCL/P risk. However, the results turned out to be different when stratified by ethnicity. BMP4 rs17563 polymorphism was associated with a higher risk of NSCL/P among Asian ethnicity (C vs T: OR = 1.33, 95% CI: 1.02-1.73; CC vs TT: OR = 2.10, 95% CI: 1.28-3.43; CC vs TT + TC: OR = 2.16, 95% CI: 1.34-3.47) and among Caucasian population (TC vs TT: OR = 3.36, 95% CI: 2.03-5.54; TC + CC vs TT: OR = 3.71, 95% CI: 2.43-5.69). Among Brazilian population, BMP4 rs17563 polymorphism exerted a significantly protective effect on NSCL/P (C vs T: OR = 0.70, 95% CI: 0.58-0.84; CC vs TT: OR = 0.54, 95% CI: 0.33-0.88; TC vs TT: OR = 0.55, 95% CI: 0.44-0.69; TC + CC vs TT: OR = 0.56, 95% CI: 0.45-0.69). CONCLUSION: The results suggest that the C allele of BMP4 rs17563 may be a risk factor for NSCL/P among Asians and Caucasians, and may be a protective factor for NSCL/P in Brazilian population. Future large-sample studies with appropriate designs among specific populations are warranted to evaluate the association.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Polymorphism, Single Nucleotide , Cleft Lip/complications , Cleft Lip/ethnology , Cleft Palate/complications , Cleft Palate/ethnology , Genetic Predisposition to Disease , Humans
11.
Medicine (Baltimore) ; 96(16): e6391, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28422830

ABSTRACT

Deep sternal wound infection is a severe complication after open heart surgery. According to the different severity and dimensions of the deep sternal wound infection, the treatment method is different. In this study, we aimed to describe our experience with the rectus abdominis myocutaneous flap for large sternal wound management, especially when 1 or 2 internal mammary arteries were absent.Between October 2010 and January 2016, a retrospective review of 9 patients who suffered from the extensive thoracic defects after deep sternal wound infection was conducted. All of these sternal defects encompassed almost the full length of the sternum after debridement. Defect reconstruction was achieved by covering with a rectus abdominis myocutaneous flap. When the ipsilateral or bilateral internal mammary artery had been harvested previously, we took advantage of the inferior epigastric artery to provide additional blood supply to the rectus abdominis myocutaneous flap. Thus, this flap had a double blood supply.There was no recurrent infection in all 9 patients. Three patients received the rectus abdominis myocutaneous flap with a double blood supply. Flap complications occurred in 2 patients (22%). One patient who did not have the double blood supply flap suffered from necrosis on the distal part of the flap, which was then debrided and reconstructed with a split-skin graft. The other patient had a seroma at the abdomen donor site and was managed conservatively. None of the patients died during the hospital stay.This study suggests that the rectus abdominis myocutaneous flap may be a good choice to repair the entire length of sternal wound. When 1 or 2 internal mammary arteries have been harvested, the inferior epigastric artery can be anastomosed to the second intercostal artery or the internal mammary artery perforator to provide the rectus abdominis myocutaneous flap with a double blood supply.


Subject(s)
Myocutaneous Flap , Rectus Abdominis/surgery , Sternum/surgery , Surgical Wound Infection/therapy , Aged , Cardiac Surgical Procedures/adverse effects , Debridement , Epigastric Arteries/surgery , Female , Humans , Male , Mammary Arteries/surgery , Middle Aged , Myocutaneous Flap/adverse effects , Myocutaneous Flap/blood supply , Myocutaneous Flap/pathology , Necrosis , Rectus Abdominis/blood supply , Rectus Abdominis/pathology , Retrospective Studies , Surgical Wound Infection/pathology
12.
Int J Mol Med ; 39(1): 153-159, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27909731

ABSTRACT

Keloids, partially considered as benign tumors, are characterized by the overgrowth of fibrosis beyond the boundaries of the wound and are regulated mainly by transforming growth factor (TGF)-ß1, which induces the transition of fibroblasts to myofibroblasts. Hypoxia is an important driving force in the development of lung and liver fibrosis by activating hypoxia inducible factor-1α and stimulating epithelial­mesenchymal transition. However, it is unknown whether and hypoxia can influence human dermal scarring. The aim of this study was to investigate whether hypoxia drives the transition of dermal fibroblasts to myofibroblasts and to clarify the potential transduction mechanisms involved. First, we observed that keloids are a relatively hypoxic tissue. Second, we found that hypoxia drives the transition of normal dermal fibroblasts to a myofibroblast-like phenotype [high expression of α-smooth muscle actin (α-SMA) and collagen I and III]. Finally, hypoxia effectively facilitated the nuclear import of the Smad2 and Smad3 complex, while blockade with the Smad3 inhibitor, SIS3, significantly impaired the expression of hypoxia-induced fibrosis-related molecules. Taken together, to the best of our knowledge, this study demonstrates for the first time that hypoxia facilitates the transition of dermal fibroblasts to myofibroblasts through the activation of the TGF-ß1/Smad3 signaling pathway and our findings may provide a potential target for the treatment of keloids.


Subject(s)
Dermis/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Cell Hypoxia , Fibrosis , Fluorescent Antibody Technique , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Keloid/pathology , Male , Phenotype , Young Adult
13.
Am J Transl Res ; 8(8): 3460-70, 2016.
Article in English | MEDLINE | ID: mdl-27648136

ABSTRACT

Keloid, a skin benign tumor, is characterized by overgrowth of fibroblasts and the excessive deposition of extracellular matrix in wounded skin. Peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist was recently evaluated to inhibit fibrosis. This study explored the underlying mechanisms. Fibroblasts isolated from 25 keloid patients (KFs) and fibroblasts isolated from healthy controls (NSFBs) were also subjected to treatment with PPAR-γ agonist troglitazone and antagonist GW9662 or for transfection with miR-92 mimics or inhibitor, Axl siRNA, and miR-92b or Axl promoter constructs, as well as being subjected to qRT-PCR, ELISA, Western blot, protein array, luciferase, and ChIP assays. The data demonstrated that TGF-ß1 and Axl proteins were significantly elevated in samples from keloid patients, while troglitazone treatment significantly reduced levels of TGF-ß1 and Axl mRNA and proteins in KFs. Moreover, knockdown of Axl expression reduced expression of TGF-ß1 and its pathway genes (such as α-SMA and Snail). PPAR-γ regulation of Axl expression was through transcriptional activation of miR-92b. miR-92b expression downregulated Axl expression at both mRNA and protein levels, whereas GW9662 completely reversed the inhibitory effects of miR-92b mimics on Axl expression. Gene ontology analysis of miR-92b targeting genes showed that TGF-ß and Axl were both potential targets of miR-92b, as confirmed by luciferase assay. These findings demonstrated that PPAR-γ-induced miR-92b expression inhibited Axl expression and in turn reduced expression of TGF-ß1 and the downstream genes in KFs, suggesting that targeting of this novel gene pathway may be useful for therapeutic control of fibrosis or keloid.

14.
Sci Rep ; 6: 32199, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27599451

ABSTRACT

Acute kidney injury (AKI) is a common complication after severe burns. Melatonin has been reported to protect against multiple organ injuries by increasing the expression of SIRT1, a silent information regulator that regulates stress responses, inflammation, cellular senescence and apoptosis. This study aimed to investigate the protective effects of melatonin on renal tissues of burned rats and the role of SIRT1 involving the effects. Rat severely burned model was established, with or without the administration of melatonin and SIRT1 inhibitor. The renal function and histological manifestations were determined to evaluate the severity of kidney injury. The levels of acetylated-p53 (Ac-p53), acetylated-p65 (Ac-p65), NF-κB, acetylated-forkhead box O1 (Ac-FoxO1), Bcl-2 and Bax were analyzed to study the underlying mechanisms. Our results suggested that severe burns could induce acute kidney injury, which could be partially reversed by melatonin. Melatonin attenuated oxidative stress, inflammation and apoptosis accompanied by the increased expression of SIRT1. The protective effects of melatonin were abrogated by the inhibition of SIRT1. In conclusion, we demonstrate that melatonin improves severe burn-induced AKI via the activation of SIRT1 signaling.


Subject(s)
Acute Kidney Injury/prevention & control , Burns/complications , Melatonin/pharmacology , Sirtuin 1/metabolism , Acetylation/drug effects , Acute Kidney Injury/etiology , Animals , Apoptosis/drug effects , Cytokines/metabolism , Kidney/drug effects , Kidney/pathology , Male , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/metabolism
15.
Am J Cancer Res ; 6(6): 1358-70, 2016.
Article in English | MEDLINE | ID: mdl-27429849

ABSTRACT

A keloid is a benign skin tumor formed by an overgrowth of granulation tissue in affected patients. Peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists were reported to be able to regulate extracellular matrix production in human dermal fibroblasts. This study explored the underlying molecular mechanism of PPAR-γ agonist troglitazone treatment for fibroblasts obtained from keloid patients. The data revealed that troglitazone treatment of keloid fibroblasts (KFs) downregulated the expression of early growth response-1 (Egr1) and collagen-1 (Col1). Level of Egr1 were closely associated with KF-induced fibrosis. The miRNA profiling data revealed that miR-543 was transcriptionally activated after troglitazone treatment. Bioinformatic analysis and experimental data showed that miR-543 was able to target Egr1. ELISA data confirmed that Col1 protein in the supernatant were modulated by the feedback regulatory axis of PPAR-γ agonist-induced miR-543 to inhibit Egr1 expression, whereas PPAR-γ antagonist treatment abolished such effect on Col1 suppression in KFs. This study demonstrated that the PPAR-γ agonist-mediated miR-543 and Egr1 signaling plays an important role in the suppression of collagen synthesis in KFs. Future in vivo studies are needed to confirm these in vitro data.

16.
Cytotherapy ; 18(8): 1012-1024, 2016 08.
Article in English | MEDLINE | ID: mdl-27262514

ABSTRACT

BACKGROUND AIMS: Keloids are raised dermal scars that extend beyond the boundaries of the initial injury. To date, there is no treatment to erase scars completely in humans. Growing evidence has shown that the human amniotic epithelial cells have anti-fibrotic properties and can reduce the fibrosis of lung and liver. However, it is unknown whether and how they can influence human keloids. The aim of this study was to investigate whether factors secreted by human amniotic epithelial cells have anti-fibrotic effects on human keloids and to clarify the potential transduction mechanism. METHODS: Human amniotic epithelial cells were isolated and identified both with flow cytometry and immunofluorescent. The α-smooth muscle actin, collagen-I and III gene and protein expression of transforming growth factor (TGF)-ß1-treated human adult dermal fibroblasts were partly abolished by human amniotic epithelial cells conditioned medium through stimulating the expression of matrix metalloproteinase (MMP). Furthermore, human amniotic epithelial cells conditioned medium effectively attenuated nuclear import of the Smad2/3 complex. RESULTS: Soluble human leukocyte antigen G, a human amniotic epithelial cell-derived factor, significantly decreased collagen production in TGF-ß1-induced human dermal fibroblasts, although the effect on collagen production was less than that of human amniotic epithelial cell-conditioned medium. CONCLUSIONS: This study demonstrates that human amniotic epithelial cells conditioned medium could down-regulate the expression of fibrosis-related molecules by regulating MMP and tissue inhibitor of metalloproteinase levels, and suppress TGF-ß1-induced fibroblast transition, in which the TGF-ß1/Smad3 pathway is likely involved. These findings suggest that human amniotic epithelial cells are a potential therapeutic compound for the treatment of keloids.


Subject(s)
Amnion/cytology , Cell Transdifferentiation/drug effects , Dermis/drug effects , Epithelial Cells/physiology , Fibroblasts/drug effects , Myofibroblasts/drug effects , Transforming Growth Factor beta1/pharmacology , Adult , Amnion/metabolism , Cells, Cultured , Collagen Type I/metabolism , Culture Media, Conditioned/pharmacology , Dermis/cytology , Dermis/physiology , Epithelial Cells/cytology , Female , Fibroblasts/physiology , Humans , Myofibroblasts/metabolism , Myofibroblasts/physiology , Pregnancy , Signal Transduction/drug effects , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
17.
Sci Rep ; 6: 24728, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27090737

ABSTRACT

Abnormally high activation of transforming growth factor-ß (TGF-ß) signaling has been demonstrated to be involved in the initiation and progression of keloids. However, the functional role of long non-coding RNA (lncRNA)-activated by TGF-ß (lncRNA-ATB) in keloids has not been documented. Here we investigated the role of lncRNA-ATB in the autocrine secretion of TGF-ß in keloid fibroblasts (KFs) and explored the underlying molecular mechanism. Using immunohistochemistry and quantitative RT-PCR analysis, we showed that lncRNA-ATB and ZNF217, a transcriptional activator of TGF-ß, were overexpressed and miR-200c, which targets ZNF217, was under-expressed in keloid tissue and keloid fibroblasts. Through gain- and loss-of-function studies, we demonstrated that knockdown of lncRNA-ATB decreased autocrine secretion of TGF-ß2 and ZNF217 expression but upregulated expression of miR-200c in KFs. Stable downregulation of ZNF217 expression decreased the autocrine secretion of TGF-ß2. miR-200c was endogenously associated with lncRNA-ATB, and inhibition of miR-200c overcame the decrease in ZNF217 expression in KFs. Taken together, these findings indicate that lncRNA-ATB governs the autocrine secretion of TGF-ß2 in KFs, at least in part, by downregulating the expression level of ZNF217 via miR-200c, suggesting a signaling axis consisting of lncRNA-ATB/miR-200c/ZNF217/TGF-ß2. These findings may provide potential biomarkers and targets for novel diagnostic and therapeutic approaches for keloids.


Subject(s)
Keloid/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Trans-Activators/genetics , Transforming Growth Factor beta2/metabolism , Biomarkers , Down-Regulation , Fibroblasts/metabolism , Gene Knockdown Techniques , Humans
18.
Sci Rep ; 6: 25272, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27121396

ABSTRACT

Recent microRNA expression profiling studies have documented an up-regulation of miR-146a in several angiogenesis models. However, the underlying molecular mechanism of miR-146a in the angiogenic activity of endothelial cells has not been clearly elucidated. The present study was aimed to evaluate whether miR-146a promotes angiogenesis in HUVECs by increasing FGFBP1 expression via directly targeting CREB3L1. miR-146a was over expressed in HUVECs via lentiviral-miR-146a. Expression profiling analysis found miR-146a over expression resulted in up-regulation of angiogenesis and cytokine activity associated genes including FGF2. Further a combination of bioinformatics and experimental analyses demonstrated the CREB3L1 as a bona fide functional target of miR-146a during angiogenesis. Moreover, CREB3L1 inhibited luciferase expression from FGFBP1 promoter containing only CRE elements. Furthermore, CREB3L1 inhibited FGFBP1 expression by binding to two CRE-like sites located at approximately -1780-1777 and -868-865 bp relative to the FGFBP1 transcription start site. Additionally, ectopic expression of CREB3L1 decreased miR-146a-induced FGF2 secretion. These findings indicate that the miR-146a-CREB3L1-FGFBP1 signaling axis plays an important role in the regulation of angiogenesis in HUVECs and provides a potential therapeutic target for anti-angiogenic therapeutics.


Subject(s)
Carrier Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Intercellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Neovascularization, Physiologic , Nerve Tissue Proteins/metabolism , Signal Transduction , Up-Regulation , Cytokines/metabolism , Humans
19.
Br J Pharmacol ; 173(10): 1589-601, 2016 05.
Article in English | MEDLINE | ID: mdl-26891034

ABSTRACT

BACKGROUND AND PURPOSE: Sirtuin1 (SIRT1), the founding member of mammalian class III histone deacetylases, is reported to be a drug target involved in fibrotic diseases. However, whether it is an effective drug target in hypertrophic scar treatment is still not known. EXPERIMENTAL APPROACH: In the present study, we observed that SIRT1 localized to both the epidermis and the dermis of skin tissues by immunohistochemistry. After knock-down of SIRT1 by shRNA or up-regulating SIRT1 by resveratrol, the expression of α-SMA, Col1 and Col3 in fibroblasts were detected by western blots. A mouse excision wound healing model was used to observe the changes in collagen fibre associated with the different expression levels of SIRT1. KEY RESULTS: SIRT1 expression was inhibited in hypertrophic scar tissue. The down-regulation of SIRT1 resulted in an increased expression of α-SMA, Col1 and Col3 in hypertrophic scar-derived fibroblasts. In contrast, the up-regulation of SIRT1 not only inhibited the expression of α-SMA, Col1 and Col3 in hypertrophic scar-derived fibroblasts but also blocked the activation of TGFß1-induced normal skin-derived fibroblasts. In the mouse model of wound healing, the deletion of SIRT1 resulted in denser collagen fibres and a more disordered structure, whereas resveratrol treatment led to a more organized and thinner collagen fibre, which was similar to that observed during normal wound healing. CONCLUSIONS AND IMPLICATIONS: The results revealed that SIRT1 negatively regulates TGFß1-induced fibroblast activation and inhibits excessive scar formation and is, therefore, a promising drug target for hypertrophic scar formation.


Subject(s)
Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/metabolism , Sirtuin 1/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , RNA, Small Interfering/genetics , Resveratrol , Sirtuin 1/biosynthesis , Sirtuin 1/deficiency , Sirtuin 1/genetics , Stilbenes/pharmacology
20.
Cell Tissue Res ; 365(1): 85-99, 2016 07.
Article in English | MEDLINE | ID: mdl-26888423

ABSTRACT

Wound healing is a highly orchestrated physiological process consisting in a complex interaction of cellular and biochemical events. Human amniotic epithelial stem cells (HAESCs) have been shown to be an attractive resource for wound healing because they are primitive stem cells. However, the exact effects of amnion-derived stem cells on the migration or proliferation of keratinocytes and their potential mechanism are not fully understood. We have found that HAESCs accelerate the migration of keratinocytes and induce a remarkable increase in the activity of phospho-ERK, phospho-JNK, and phospho-AKT, the blockade of which by their specific inhibitors significantly inhibits migration induced by HAESC-conditioned medium (CM). Furthermore, the co-culture of keratinocytes with HAESCs up-regulates the expression levels of cell proliferation proteins Cyclin D1, Cyclin D3 and Mdm2. In vivo animal experiments have shown that HAESC-CM improves wound healing, whereas blockade with ERK, JNK and AKT inhibitors significantly impairs wound healing. Taken together, these results reveal, for the first time, that HAESCs promote wound healing by facilitating the migration and proliferation of keratinocytes via ERK, JNK and AKT signaling pathways and might be a potential therapy in skin wound healing.


Subject(s)
Amnion/cytology , Cell Movement , Epithelial Cells/cytology , Keratinocytes/cytology , Keratinocytes/enzymology , Signal Transduction , Stem Cells/cytology , Wound Healing , Adult , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Separation , Coculture Techniques , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Keratinocytes/drug effects , Male , Mice, Inbred C57BL , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Skin/drug effects , Skin/pathology , Stem Cells/drug effects , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...