Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Med Rep ; 29(2)2024 Feb.
Article in English | MEDLINE | ID: mdl-38099341

ABSTRACT

Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that the western blotting data shown in Figs. 4B and 5 and the H&E immunostaining data shown in Fig 1A were strikingly similar to data appearing in different form in other articles written by different authors at different research institutes that had either already been published, or were submitted for publication at around the same time.  Owing to the fact that the contentious data in the above article had already been published prior to its submission to Molecular Medicine Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Molecular Medicine Reports 13: 2683-2688, 2016; DOI: 10.3892/mmr.2016.4877].

2.
3.
Exp Ther Med ; 22(4): 1135, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34466145

ABSTRACT

[This retracts the article DOI: 10.3892/etm.2017.4595.].

4.
Oncol Rep ; 46(4)2021 Oct.
Article in English | MEDLINE | ID: mdl-34368877

ABSTRACT

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that certain of the flow cytometric data shown in Fig. 3, and western blotting assay data shown in Fig. 6F, were strikingly similar to data appearing in different form in other articles by different authors. Owing to the fact that the contentious data in the above article had already been published elsewhere, or were already under consideration for publication, prior to its submission to Oncology Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive any reply. The Editor apologizes to the readership for any inconvenience caused. [the original article was published in Oncology Reports 36: 471­479, 2016; DOI: 10.3892/or.2016.4824].

5.
Exp Ther Med ; 14(6): 5611-5618, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29285100

ABSTRACT

Cutaneous squamous cell carcinoma (CSCC) is the second most common type of skin cancer with increasing incidence. In recent years, several microRNAs (miRs) have been demonstrated to serve an oncogenic or tumor suppressive role in CSCC. However, the exact role of miR-34a in CSCC and the underlying regulatory mechanism remain unclear. The present study aimed to investigate the regulatory mechanism of miR-34a in the malignant phenotypes of CSCC cells using MTT assay, wound healing assay and transwell assay. It was observed that miR-34a was significantly downregulated in CSCC tissues and cell lines, and low miR-34a expression was associated with the aggressive progression of CSCC. Restoration of miR-34a significantly suppressed the proliferation, migration and invasion of CSCC SCL-1 cells. High-mobility group box 1 (HMGB1) was then identified as a target gene of miR-34a in SCL-1 cells using bioinformatics prediction. The expression of HMGB1 was significantly upregulated in the CSCC tissues and cell lines. Furthermore, the protein expression of HMGB1 was negatively regulated by miR-34a in SCL-1 cells, while overexpression of HMGB1 impaired the inhibitory effects of miR-34a on SCL-1 cells. These findings suggest that miR-34a represses the malignant phenotypes of CSCC cells, at least partly, via the inhibition of HMGB1. Therefore, miR-34a may be used as a promising therapeutic candidate for CSCC.

6.
Exp Ther Med ; 14(2): 931-938, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28810544

ABSTRACT

MicroRNA (miR) are a class of small non-coding RNA that are able to inhibit gene expression by directly binding to the 3' untranslated region (UTR) of their target mRNA and thus promote translational repression or mRNA degradation. Recently, miR-9 was reported to have a suppressive role in malignant melanoma; however, the underlying mechanism remains largely unclear. In the present study, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were used to examine the mRNA and protein expression levels in malignant melanoma tissues and cell lines. The MTT assay and wound healing assay were used to examine the cell viability, proliferation and migratory capacities. Bioinformatics prediction and luciferase reporter assay were performed to investigate the relationship between miR-9 and its potential target gene. The present data revealed that miR-9 expression was significantly downregulated in malignant melanoma tissues when compared with their matched adjacent non-tumor tissues. Furthermore, the expression levels of miR-9 were reduced in malignant melanoma cell lines when compared with human normal skin HACAT cells. Moreover, the ectopic expression of miR-9 significantly suppressed the proliferation and migration of malignant melanoma cells, accompanied with a remarkable decrease in the protein expression levels of sirtuin 1 (SIRT1), which were markedly upregulated in malignant melanoma tissues and cell lines. Additionally, restoration of SIRT1 reversed the suppressive effects of miR-9 on the proliferation and migration of malignant melanoma cells. Luciferase reporter assay data further identified SIRT1 as a direct target gene of miR-9. To conclude, the present findings indicate that miR-9 has a suppressive role in malignant melanoma cell viability and migration, at least in part, via directly inhibiting the protein expression of its target gene, SIRT1. Therefore, miR-9 may serve as a potential candidate for the treatment of malignant melanoma.

7.
J Craniofac Surg ; 28(6): 1418-1424, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28692513

ABSTRACT

OBJECTIVES: The objective of this study was to investigate the viability and biomechanics of diced cartilage blended with platelet-rich plasma (PRP) and wrapped with poly (lactic-co-glycolic) acid (PLGA) membrane in a rabbit model. METHODS: A total of 10 New Zealand rabbits were used for the study. Cartilage grafts were harvested from 1 side ear. The grafts were divided into 3 groups for comparison: bare diced cartilage, diced cartilage wrapped with PLGA membrane, and diced cartilage blended with PRP and wrapped with PLGA membrane. Platelet-rich plasma was prepared using 8 mL of auricular blood. Three subcutaneous pockets were made in the backs of the rabbits, and the grafts were placed in these pockets. The subcutaneous implant tests were conducted for safety assessment of the PLGA membrane in vivo. All of the rabbits were sacrificed at the end of 3 months, and the specimens were collected. The sections were stained with hematoxylin and eosin, toluidin blue, and collagen II immunohistochemical. Simultaneously, biomechanical properties of grafts were assessed. RESULTS: This sample of PLGA membrane was conformed to the current standard of biological evaluation of medical devices. Moderate resorption was seen at the end of 3 months in the gross assessment in diced cartilage wrapped with PLGA membrane, while diced cartilage blended with PRP had no apparent resorption macroscopically and favorable viability in vivo after 3 months, and the histological parameters supported this. Stress-strain curves for the compression test indicated that the modulus of elasticity of bare diced cartilage was 7.65 ±â€Š0.59 MPa; diced cartilage wrapped with PLGA membrane was 5.98 ±â€Š0.45 MPa; and diced cartilage blended with PRP and wrapped with PLGA membrane was 7.48 ±â€Š0.55 MPa, respectively. CONCLUSIONS: Diced cartilage wrapped with PLGA membrane had moderate resorption macroscopically after 3 months. However, blending with PRP has beneficial effects in improving the viability of diced cartilages. Additionally, the compression modulus of diced cartilage blended with PRP and wrapped with PLGA membrane was similar to bare diced cartilage.


Subject(s)
Cartilage , Lactic Acid/pharmacology , Platelet-Rich Plasma , Polyglycolic Acid/pharmacology , Tissue Survival , Animals , Cartilage/drug effects , Cartilage/physiology , Elastic Modulus , Membranes, Artificial , Polylactic Acid-Polyglycolic Acid Copolymer , Rabbits , Tissue Survival/drug effects , Tissue Survival/physiology
8.
Onco Targets Ther ; 9: 7047-7057, 2016.
Article in English | MEDLINE | ID: mdl-27895497

ABSTRACT

MicroRNAs (miRs) are a class of small noncoding RNAs that negatively regulate the gene expression by directly binding to the 3' untranslated region of their target mRNA, thus resulting in mRNA degradation or translational repression. miR-9 has recently been demonstrated to play a role in the development and progression of malignant melanoma (MM), but the regulatory mechanism of miR-9 in the malignant phenotypes of MM still remains largely unknown. In this study, a total of 73 pairs of MM tissues and adjacent normal tissues were collected. Real-time reverse transcription polymerase chain reaction and Western blot were used to detect the mRNA and protein expression of miR-9. MTT assay, wound healing assay, and transwell assay were conducted to determine the cell proliferation, migration, and invasion. Luciferase reporter assay was used to determine the targeting relationship between miR-9 and NRP1. Our data demonstrated that miR-9 expression was significantly downregulated in MM tissues compared with that in adjacent normal tissues. The decreased miR-9 level was significantly associated with the tumor stage and metastasis of MM. We also found that the expression level of miR-9 was decreased in MM cell lines (G361, B16, A375, and HME1) compared with normal skin HACAT cells. Ectopic expression of miR-9 led to a significant decrease in the ability of proliferation, migration, and invasion in A375 cells. NRP1 was further identified as a direct target gene of miR-9, and the protein expression of NRP1 was negatively regulated by miR-9 in A375 cells. Furthermore, overexpression of NRP1 reversed the suppressive effects of miR-9 on the malignant phenotypes of A375 cells. In vivo study revealed that miR-9 overexpression decreased the tumor growth, while overexpression of NRP1 increased MM growth. In summary, our findings suggest that the miR-9/NRP1 axis may serve as a potential target for the treatment of MM.

9.
Clin Exp Pharmacol Physiol ; 43(10): 921-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27385584

ABSTRACT

The aim of this study was to determine whether systemic inflammatory response syndrome (SIRS) in burn patients is mediated by the brain natriuretic peptide (BNP)/natriuretic peptide A receptor (NPRA)-induced heat shock factor 1 (HSF-1) signalling pathway. Mononuclear cells (MNCs) that were isolated from patients with burn injuries and SIRS mouse models and a RAW264.7 cell line were treated with normal serum or serum obtained from animals with burn injuries. In parallel, small hairpin RNAs (shRNAs) against BNP or NPRA were transfected in both cell types. Western blotting (WB) and enzyme-linked immunosorbent assay (ELISA) were used to detect protein expression and inflammatory factor levels, respectively. We found that interleukin (IL)-12, tumour necrosis factor (TNF)-α, C-reactive protein (CRP), and BNP levels were increased and IL-10 levels were decreased in the plasma and MNCs in vivo in the animal model of SIRS. Additionally, NPRA was upregulated, whereas HSF-1 was downregulated in monocytes in vivo. Treatment of RAW264.7 cells with burn serum or BNP induced IL-12, TNF-α, and CRP secretion as well as HSF-1 expression. Finally, silencing BNP with shRNA interrupted the effect of burn serum on RAW264.7 cells, and silencing NPRA blocked burn serum- and BNP-mediated changes in RAW264.7 cells. These results suggest that the interaction of NPRA with BNP secreted from circulatory MNCs as well as mononuclear macrophages leads to inflammation via HSF-1 during SIRS development following serious burn injury.


Subject(s)
Burns/blood , DNA-Binding Proteins/blood , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, C-Type/blood , Protein Precursors/blood , Systemic Inflammatory Response Syndrome/blood , Transcription Factors/blood , Animals , Atrial Natriuretic Factor , Biomarkers/blood , Burns/complications , Cell Line , Heat Shock Transcription Factors , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Systemic Inflammatory Response Syndrome/etiology
10.
Oncol Rep ; 36(1): 471-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27220837

ABSTRACT

MicroRNAs (miRs) have been demonstrated to play critical roles in the development and progression of malignant melanoma (MM). However, the exact role and underlying mechanism of miR-18b in MM growth remains unclear. In the present study, real-time PCR data indicated that miR-18b was significantly downregulated in MM tissues compared to their matched adjacent non-tumor tissues. Low miR-18b expression was significantly associated with the tumor thickness and stage, although no significant association was observed between the miR-18b expression and the age, gender, or lymph node metastasis. Besides, miR-18b was also significantly downregulated in MM B16 and A375 cells compared to normal skin HACAT cells. Ectopic expression of miR-18b decreased the proliferation of A375 and B16 cells, while induced a remarkable cell cycle arrest at G1 stage. Besides, miR-18b overexpression also inhibited the glycolysis in A375 and B16 cells. HIF-1α, a key regulator in glycolysis, was then identified as a target gene of miR-18b, and its expression was negatively mediated by miR-18b in A375 and B16 cells. Overexpression of HIF-1α rescued the suppressive effect of miR-18b on MM cell proliferation and glycolysis. In vivo study further showed that overexpression of miR-18b inhibited the MM growth as well as the tumor-related death, accompanied with HIF-1α downregulation. Taken together, the present study suggests that miR-18b inhibits the growth of MM cells in vitro and in vivo through directly targeting HIF-1α.


Subject(s)
Cell Proliferation/genetics , Glycolysis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Melanoma/genetics , MicroRNAs/genetics , Cell Cycle Checkpoints/genetics , Cell Line , Cell Line, Tumor , Down-Regulation/genetics , Female , G1 Phase/genetics , HEK293 Cells , Humans , Lymphatic Metastasis/genetics , Male , Melanoma, Experimental/genetics , Middle Aged , Signal Transduction/genetics
11.
Mol Med Rep ; 13(3): 2683-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26861712

ABSTRACT

MicroRNAs (miRs), a class of non­coding RNAs 18­25 nucleotides in length, generally serve suppressive role in the regulation of gene expression via directly binding to the 3'­untranslated region (UTR) of their target mRNA. Previous studies have identified several miRs to be involved in thermal injury repair. However, the role of miR let­7b during the recovery of thermal injury, in addition to the underlying mechanisms, has not previously been studied. In the present study, the expression of let­7b was observed to be significantly increased in skin tissue shortly following thermal injury, however, gradually reduced during the recovery of thermal injury. Notably, similar findings were observed in heat­denatured skin fibroblasts. Furthermore, collagen, type I, alpha 1 (COL1A1) and collagen, type I, alpha 2 (COL1A2), which are associated with the synthesis of type I collagen, were identified as two targets of let­7b in skin fibroblasts. The overexpression of let­7b was observed to upregulate the protein expression levels of COL1A1 and COL1A2, while knockdown of let­7b reduced the levels of COL1A1 and COL1A2 in skin fibroblasts. Furthermore, COL1A1 and COL1A2 were significantly downregulated shortly following thermal injury, while gradually upregulated during healing, in heat­damaged skin tissue and skin fibroblasts, with the expression profiles opposite to that of let­7b. Taken together, this suggests that the downregulation of let­7b in heat­damaged dermis promotes the synthesis of type I collagen and thus aids in burn wound repair.


Subject(s)
Burns/metabolism , Collagen Type I/metabolism , MicroRNAs/metabolism , Skin/metabolism , Wound Healing , Animals , Cell Line , Collagen Type I, alpha 1 Chain , Dermis/metabolism , Down-Regulation , Fibroblasts/metabolism , Humans , Male , Rats, Sprague-Dawley
12.
Medicine (Baltimore) ; 94(7): e458, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25700309

ABSTRACT

The aim of the present study was to compare expression of microRNAs (miRNAs) from scar and normal skin areas in patients who suffered acute injuries in the skin. A total of 9 patients with acute injuries in the skin who received surgical treatment from December 2012 to March 2013 were included in this pilot study. Specimens from the hypertrophic scar and normal skin areas were obtained from the same patient during surgery. To screen for differentially expressed miRNAs, we applied 3 statistical methods, namely the traditional t test, the false discovery rate (FDR), and a novel sure independence screening procedure based on the distance correlation (DC-SIS). We examined the functional trends and metabolic and regulatory pathways for the target genes of the identified miRNAs, and explored interaction of these miRNAs in the implication of scar healing using Ingenuity Pathway Analysis. DC-SIS identified 18 differentially expressed miRNAs, 4 of which (miR-149, miR-203a, miR-222, miR-122) were also identified by FDR. The target genes of the 4 miRNAs exhibit a variety of biological functions, and are involved in various pathways such as mitogen-activated protein kinase, Wnt signaling, and focal adhesion. We identified 1 network in which 14 out of the 18 differentially expressed miRNAs were involved. Many of the miRNAs in the network target genes were involved in cell proliferation and apoptosis.In this pilot study, we identified several miRNAs exhibiting differential expression in patients who suffered acute injuries in the skin. Further studies on these miRNAs are needed to validate our findings and explore their roles in the wound healing process of the skin.


Subject(s)
Cicatrix/genetics , Cicatrix/physiopathology , MicroRNAs/biosynthesis , Wound Healing/genetics , Wound Healing/physiology , Adolescent , Adult , Cell Physiological Phenomena/genetics , Cell Physiological Phenomena/physiology , Child , Child, Preschool , Female , Humans , Male , Pilot Projects , Skin/physiopathology , Young Adult
13.
J Cancer Res Clin Oncol ; 141(6): 993-1006, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25403318

ABSTRACT

PURPOSE: To confirm whether flotillin 2 (FLOT2) is a direct target of miR-34a and miR-34a/FLOT2 pathway plays a key role in melanoma proliferation and metastasis. METHODS: First, miR-34a and FLOT2 expressions were both detected in human tissues and cell lines by qRT-PCR. Then, after transfection of mimics/inhibitor of miR-34a into melanoma cell lines, MTT, colony formation, scratch migration assays and transwell invasion assays were performed to evaluate the impact of miR-34a on cell proliferation and metastasis. Western blot, qRT-RCR and dual luciferase reporter gene assays were carried out to confirm whether FLOT2 is a direct target gene of miR-34a. In functional recovery experiments, proliferation and metastasis ability of WM35 and WM451 was tested after being co-transfected with miR-34a inhibitor/si-FLOT2 or miR-34a mimics/FLOT2 cDNA to confirm that FLOT2 is downregulated by miR-34a. RESULTS: The miR-34a significantly lower-expressed in metastasis melanoma tissues compared to in situ melanoma, nevi and normal skin whereas FLOT2 has an opposite trend. The level of miR-34a and FLOT2 in different melanoma cell lines was also tested and found that metastatic melanoma cell lines has lower miR-34a expression and higher FLOT2 expression compare to in situ melanoma cell line. MiR-34a overexpression profoundly inhibits WM451 cell proliferation and metastasis, whereas miR-34a reduction had a promoting effect to proliferation and metastasis of WM35. Results of Western blot, qRT-RCR and dual luciferase reporter gene assays revealed that FLOT2 is a direct target gene of miR-34a. Furthermore, overexpression/blockage of FLOT2 could attenuate effect of miR-34a overexpression/inhibition which indicated miR-34a suppresses melanoma biological behavior partially through FLOT2 inhibition. CONCLUSIONS: Our study confirmed that miR-34a is involved in the tumor inhibition of melanoma by directly targeting FLOT2 gene. This finding provides potential novel strategies for therapeutic interventions of melanoma.


Subject(s)
Melanoma/drug therapy , Melanoma/pathology , Membrane Proteins/metabolism , MicroRNAs/metabolism , Molecular Targeted Therapy , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/metabolism , Membrane Proteins/drug effects , MicroRNAs/pharmacology , Molecular Targeted Therapy/methods , Real-Time Polymerase Chain Reaction , Skin Neoplasms , Up-Regulation/drug effects , Melanoma, Cutaneous Malignant
14.
Int J Clin Exp Pathol ; 7(10): 7131-41, 2014.
Article in English | MEDLINE | ID: mdl-25400809

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC), the second most common form of human cancer, is an epithelial skin tumor, which can result in metastasis with lethal consequences accounting for about 20% of all skin cancer-related deaths. The metastasis is the main reason for cSCC-related deaths with an overall 5-year survival rate < 30% in cases that spread systemically. The role of miRNAs has been involved in SCC of different origins. Recent data have revealed that the expression of miRNA-199a was changed in many human cancers. In this study, we found that miR-199a was significantly decreased in cSCC tissues, which had an inverse relationship with CD44. MiR-199a specifically regulated the expression of CD44 at mRNA and protein levels, and the interaction between CD44 and Ezrin in cSCC cells. Moreover, the suppressive role of miR-199a in cell migration in cSCC cells was also associated with the activity of MMP2 and MMP9. Taken together, our data indicated that increased expression of endogenous mature miR-199a might prevent the growth and migration of human cSCC via decreasing the expression of CD44 and regulating the interaction between CD44 and Ezrin, which may provide a potentially important therapeutic target for human cSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Movement , Cell Proliferation , Cytoskeletal Proteins/metabolism , Hyaluronan Receptors/metabolism , MicroRNAs/metabolism , Skin Neoplasms/metabolism , 3' Untranslated Regions , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , MicroRNAs/genetics , Neoplasm Invasiveness , RNA, Messenger/metabolism , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Time Factors , Transfection
15.
J Dermatol ; 41(10): 903-11, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25228082

ABSTRACT

Hypertrophic scarring leads to a deformed appearance and contracted neogenetic tissue, resulting in physiological and psychological problems for patients. Millions of people suffer these discomforts each year. Emerging evidence has reported that miRNA contributed to hypertrophic scarring or keloid formation. In this study, nine hypertrophic scar samples and the matched normal skin tissues were used to perform a miRNA microarray. The results of miRNA array showed that miR-200b was downregulated by more than 2-fold, validated by qPCR in hypertrophic scar tissues and human hypertrophic scar fibroblasts, suggesting that there was an important correlation between miR-200b and hypertrophic scarring. We also found that miR-200b affected hypertrophic scarring through regulating the cell proliferation and apoptosis of human hypertrophic scar fibroblasts by affecting the collagen I and III synthesis, fibronectin expression and TGF-ß1/α-SMA signaling. Thus, our study provides evidence to support that miR-200b may be a useful target for hypertrophic scarring management.


Subject(s)
Apoptosis , Cell Proliferation , Cicatrix, Hypertrophic/metabolism , Fibroblasts/metabolism , MicroRNAs/metabolism , Actins/metabolism , Adolescent , Adult , Caspase 3/metabolism , Caspase 8/metabolism , Child , Child, Preschool , Cicatrix, Hypertrophic/etiology , Collagen Type I/biosynthesis , Collagen Type III/biosynthesis , Female , Fibronectins/metabolism , Gene Expression Profiling , Humans , Male , Oligonucleotide Array Sequence Analysis , Proliferating Cell Nuclear Antigen/metabolism , Transforming Growth Factor beta1/metabolism , Young Adult
16.
Cancer Biol Ther ; 15(10): 1340-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25019203

ABSTRACT

BACKGROUND: MicroRNA-20a (miR-20a) plays a key role in tumorigenesis and progression. But its function is reverse in different kinds of malignant tumor, and its role and mechanism in cutaneous squamous cell carcinoma (CSCC) remains unclear. OBJECT: To determine the miR-20a's roles in CSCC and confirm whether LIMK1 is a direct target gene of miR-20a. METHODS: First miR-20a and LIMK1 expression levels were detected in six pairs of CSCC tissues and corresponding normal skin by qRT-PCR. Then MTT assays and colony formation assays were performed to evaluate the impact of miR-20a on cell proliferation. In addition, scratch migration assays and transwell invasion assays were performed to check miR-20a's effect on cell metastasis. Since LIMK1 (LIM kinase-1) was predicted as a target gene of miR-20a, the changes of LIMK1 protein and mRNA were measured by western blot and qRT-RCR methods after miR-20a overexpression. Moreover the dual reporter gene assay was performed to confirm whether LIMK1 is a direct target gene of miR-20a. Finally LIMK1 mRNA and miR-20a in other 30 cases of CSCC pathological specimens were determined and a correlation analysis was evaluated. RESULTS: The miR-20a significantly low-expressed in CSCC tissues compared with that in matched normal tissues while LIMK1 has a relative higher expression. MiR-20a inhibited A431 and SCL-1 proliferation and metastasis. Both of LIMK1 protein and mRNA levels were downregulated after miR-20a overexpression. The dual reporter gene assays revealed that LIMK1 is a direct target gene of miR-20a. Furthermore, qRT-PCR results of LIMK1 mRNA and miR-20a in 30 cases of CSCC pathological specimens showed miR-20a is inversely correlated with LIMK1 expression. CONCLUSION: Our study demonstrated that miR-20a is involved in the tumor inhibition of CSCC by directly targeting LIMK1 gene. This finding provides potential novel strategies for therapeutic interventions of CSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Proliferation , Lim Kinases/metabolism , MicroRNAs/metabolism , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/secondary , Cell Movement , Female , Humans , Lim Kinases/genetics , MicroRNAs/genetics , Tumor Cells, Cultured
17.
Exp Ther Med ; 6(6): 1390-1396, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24255668

ABSTRACT

Burn injuries are common in wartime and in times of peace. The prevention and therapy of ischemia-reperfusion injury to the organs, in particular the intestine, during the burn shock and recovery process has become a popular yet challenging area of research. Studies concerning the apoptosis of the cells of the burned intestinal mucosa have gained considerable attention. Qinghuobaiduyin (QHBDY) is a traditional Chinese medicine that has been used as a clinical prescription since 1995 to treat burn patients due to its opsonization function in the immune system and favorable clinical therapeutic effect. The aim of this study was to investigate the effect of QHBDY on the apoptosis of intestinal mucosa following burn injury. An animal model was constructed comprising severely burned rats that were treated with various dosages of QHBDY. Tissues from the small intestine were collected to investigate the apoptosis rate by TUNEL assay and the protein expression levels of heat shock protein 70 (Hsp70) and caspase-3 by immunohistochemistry. In addition, IEC-18 cells treated with QHBDY and burn serum were investigated. The cell apoptosis rate was analyzed by flow cytometry (FCM), the protein expression levels of Hsp70 were measured by western blot analysis and caspase-3 activity was analyzed by a colorimetric assay. The results showed that in animal experiments, compared with the burned group, the apoptosis rates in the treatment group was decreased, the protein expression level of Hsp70 was increased while Caspase-3 was decreased. In cell experiments, after treatment with QHBDY, the cell apoptosis rate was lower than that of the burn serum group. In addition, Hsp70 protein expression was upregulated and caspase-3 activity was decreased. QHBDY may play an important role in the prevention of apoptosis at the whole animal and cellular levels.

18.
Exp Ther Med ; 6(1): 223-227, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23935750

ABSTRACT

The aim of this study was to explore the etiology and diagnosis of multiple intracranial hemorrhages (ICHs) following severe burns, with a retrospective review of 16 cases of severe burns further complicated by multiple ICHs. Using cranial CT scans of the brains, we identified that all patients presented with low platelet counts and coagulation abnormalities prior to intracranial hemorrhaging. Following conventional treatment and various supporting treatments, five cases succumbed following a progressive reduction in blood platelet levels and the ICHs were cured in 11 cases following the restoration of normal platelet levels. We conclude that low platelet counts and coagulation abnormalities may cause multiple ICHs following severe burns and early diagnosis and treatment is the key to successful treatment.

19.
J Surg Res ; 185(1): 319-28, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23866790

ABSTRACT

BACKGROUND: High-mobility group box 1 protein (HMGB1), a downstream inflammatory response modifier in sepsis and endotoxemia, alters endotoxin tolerance by affecting cellular hyporesponsiveness and tumor necrosis factor α and interleukin 1 production. OBJECTIVE: Endogenous HMGB1 signaling mechanisms during low-dose lipopolysaccharide (LPS)-induced endotoxin tolerance were investigated. METHODS: BALB/c mice were preconditioned with either 0.1 mL low-dose LPS (0.2 mg/kg) or phosphate-buffered saline (PBS) (control) followed by treatment with three consecutive injections of anti-HMGB1, IgY (an nonspecific antibody), or PBS, at 2, 12, and 22 h, respectively, Mice were then subjected to 0.1 mL high-dose LPS (10 mg/kg) or PBS at 24 h. Serum and hepatic tissue samples were obtained 1 or 3 h after final treatments. Signaling mechanisms were further investigated in the serum and hepatic tissues of mice preconditioned with 0.1 mL HMGB1 (1 mg/kg), low-dose LPS (0.2 mg/kg), or PBS for 1 h, and then high-dose LPS treatment for 3 h. RESULTS: The signaling mechanisms involved in low-dose LPS preconditioning required enhanced endogenous HMGB1 expression and secretion. Neutralizing endogenous HMGB1 with anti-HMGB1 antibodies following low-dose LPS preconditioning altered endotoxin tolerance by increasing serum tumor necrosis factor α, reducing hepatic interleukin-1R-associated kinase M expression, and partially restoring nuclear factor κB in vivo. The translocation from nucleus to cytoplasm of endogenous HMGB1 in RAW264.7 cells was also observed during low-dose LPS-induced endotoxin tolerance. CONCLUSIONS: Increased interleukin-1R-associated kinase M and decreased nuclear factor κB activity in endotoxin tolerance is associated with endogenous HMGB1 expression after low-dose LPS preconditioning. These findings provide a basis for a better mechanistic understanding and the development of safer clinical therapeutics utilizing induced endotoxin tolerance.


Subject(s)
Endotoxemia/immunology , HMGB1 Protein/immunology , Immune Tolerance/immunology , Sepsis/immunology , Signal Transduction/immunology , Animals , Autoantibodies/immunology , Cell Line , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/metabolism , HMGB1 Protein/metabolism , HMGB1 Protein/pharmacology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-1/immunology , Interleukin-1/metabolism , Interleukin-1 Receptor-Associated Kinases/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Sepsis/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
Exp Ther Med ; 5(5): 1345-1350, 2013 May.
Article in English | MEDLINE | ID: mdl-23737876

ABSTRACT

Saikosaponin A (SSA) is a major triterpenoid saponin isolated from Radix bupleuri (RB), a widely used Chinese traditional medicine to treat various inflammation-related diseases. The aim of this study was to investigate the anti-inflammatory activity, as well as the molecular mechanism of SSA in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. In this study, we demonstrated that SSA markedly inhibits the expression of certain immune-related cytotoxic factors, including cyclooxygenase-2 (COX-2) and inducible nitric-oxide synthase (iNOS), as well as pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and IL-6. It also significantly upregulates the expression of IL-10, an important anti-inflammatory cytokine, suggesting its anti-inflammatory activity in LPS-stimulated macrophages. We further demonstrated that SSA inhibits the activation of the nuclear factor-κB (NF-κB) signaling pathway by suppressing the phosphorylation of inhibitory NF-κB inhibitor α (IκBα) and thus holding p65 NF-κB in the cytoplasm to prevent its translocation to the nucleus. In addition, SSA also inhibits the mitogen-activated protein kinase (MAPK) signaling pathway by downregulating the phosphorylation of p38 MAPK, c-Jun N-terminal kinase (c-JNK) and extracellular signal-regulated kinase (ERK), the three key components of the MAPK family. In conclusion, our study demonstrates that SSA has an anti-inflammatory effect by regulating inflammatory mediators and suppressing the MAPK and NF-κB signaling pathways in LPS-stimulated RAW 264.7 cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...