Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Regen Ther ; 24: 651-661, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38074191

ABSTRACT

Background aims: Spinal cord injury (SCI) is one of the most complex and destructive diseases of the nervous system, which can lead to permanent loss of tactile perception. But existing treatment methods have limited effects. To establish a novel method that may be therapeutic in repairing the injured spinal cord, gene-modified dental pulp stem cells (DPSCs) were injected in situ. Methods: Adenovirus carrying osteopontin (OPN), Insulin-like growth factor 1 (IGF-1) and cailiary-derived neurotrophic factor (CNTF) (Ad-OIC) was constructed. After modified with Ad-OIC, supernatant of DPSC were co-cultured with HT-22 cells and the effect of DPSC-OIC on the HT-22 cells were evaluated via Cell Counting Kit-8 (CCK-8) assay, Real-Time polymerase chain reaction (PCR) analysis, laser confocal microscopy and fluorescence activating cell sorter (FACS). DPSC-OIC were injected in the lesion area of injured spinal cord and the survival time of transplanted cells were measured by bioluminescence imaging system. The recovery of the injured spinal cord was evaluated by behavioral score, radiological evaluation and immunopathological analysis. Results: DPSC-OIC could enhance the proliferation and axon growth of HT-22 cells, and protect HT-22 cells from H2O2 induced apoptosis. The transplanted DPSC-Null or DPSC-OIC could survive for more than two weeks in local injection site. DPSC-OIC treatment could increase Basso-Mouse Scale (BMS) scores, improve Magnetic Resonance Imaging (MRI) manifestation and promote bladder function recovery. Less apoptotic neurons and more proliferative cells were found in the lesion area of DPSC-OIC treated spinal cord. Nestin+ cells and neural stem cell marker (Sox2) were both up-regulated after DPSC-OIC treatment. Additionally, inhibitory extracellular matrix proteoglycan Neural/Glial Antigen 2 (NG2) was down-regulated and axon growth promotive factor fibronectin was up-regulated after both DPSC-Null (DPSCs infected with Ad-Null) and DPSC-OIC treatments. Conclusions: DPSC-OIC could be a novel effective method for treating SCI.

2.
Stem Cells Int ; 2022: 7590337, 2022.
Article in English | MEDLINE | ID: mdl-36299466

ABSTRACT

Intervertebral disc (IVD) degeneration is the primary cause for low back pain that has a high prevalence in modern society and poses enormous economic burden on patients. Few effective therapeutic strategies are available for IVD degeneration treatment. To understand the biological effects of dental pulp stem cells (DPSCs) on nucleus pulposus (NP) cells, we carried out RNA sequencing, bioinformatic analysis which unveiled gene expression differences, and pathway variation in primarily isolated patients' NP cells after treatment with DPSCs supernatant. Western blot and immunofluorescence were used to verify these molecular alterations. Besides, to evaluate the therapeutic effect of DPSCs in IVD degeneration treatment, DPSCs were injected into a degeneration rat model in situ, with treatment outcome measured by micro-CT and histological analysis. RNA sequencing and in vitro experiments demonstrated that DPSCs supernatant could downregulate NP cells' inflammation-related NF-κB and JAK-STAT pathways, reduce IL-6 production, increase collagen II expression, and mitigate apoptosis. In vivo results showed that DPSCs treatment protected the integrity of the disc structure, alleviated extracellular matrix degradation, and increased collagen fiber expression. In this study, we verified the therapeutic effect of DPSCs in an IVD degeneration rat model and elucidated the underlying molecular mechanism of DPSCs treatment, which provides a foundation for the application of DPSCs in IVD degeneration treatment.

3.
Mol Ther Oncolytics ; 24: 486-496, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35229027

ABSTRACT

Oncolytic adenoviruses (OAds) are alternative immune therapeutic strategies for tumors. However, liver uptake and antibody neutralization are two major barriers for systemic delivery during the treatment of tumor metastasis. Mesenchymal stem cells (MSCs) have emerged as potential vehicles to improve delivery. In this study, we loaded umbilical-cord-derived MSCs (UC-MSCs) with OAds expressing decorin (rAd.DCN) or without foreign genes (rAd.Null) to treat breast cancer lung metastasis. In vivo, rAd.Null, MSCs.Null, and rAd.DCN exhibited antitumor effects compared with other groups in a mouse model. Unexpectedly, MSCs.Null showed much greater antitumor responses than MSCs.DCN, including improved survival and reduced tumor burden. Compared with rAd.Null, both MSCs.Null and MSCs.DCN could improve the viral spread and distribution in metastatic tumor lesions in the lung. MSCs.DCN produced much more decorin in lungs than rAd.DCN; however, rAd.DCN reduced the downstream target genes of decorin much more strongly than MSCs.DCN, which was consistent with in vitro findings. In addition, rAd.DCN, MSCs.Null, and MSCs.DCN could reduce The cytokine levels in the lung. In conclusion, MSCs improved oncolytic adenoviral delivery and spread in tumor tissues and enhanced therapeutic effects. However, MSCs.DCN reduced OAd-evoked antitumor responses, possibly via a contact-dependent mechanism.

4.
Cancer Gene Ther ; 29(5): 456-465, 2022 05.
Article in English | MEDLINE | ID: mdl-34561555

ABSTRACT

Effective therapeutic strategies for triple-negative breast cancer (TNBC) are still lacking. Clinical data suggest that a large number of TNBC patients cannot benefit from single immune checkpoint inhibitor (ICI) treatment due to the immunosuppressive tumour microenvironment (TME). Therefore, combination immunotherapy is an alternative approach to overcome this limitation. In this article, we combined two kinds of oncolytic adenoviruses with ICIs to treat TNBC in an orthotopic mouse model. Histopathological analysis and immunohistochemistry as well as multiplex immunofluorescence were used to analyse the TME. The immunophenotype of the peripheral blood and spleen was detected by using flow cytometry. Oncolytic adenovirus-mediated immune activity in a coculture system of lytic supernatant and splenocytes supported the study of the mechanism of combination therapy in vitro. Our results showed that the combination of oncolytic adenoviruses with anti-programmed cell death-ligand 1 (anti-PD-L1) and anti-cytotoxic T lymphocyte-associated antigen-4 (anti-CTLA-4) (aPC) can significantly inhibit tumour growth and prolong survival in a TNBC model. The combination therapy synergistically enhanced the antitumour effect by recruiting CD8+ T and T memory cells, reducing the number of regulatory T cells and tumour-associated macrophages, and promoting the polarization of macrophages from the M2 to the M1 phenotype to regulate the TME. The rAd.GM regimen performed better than the rAd.Null treatment. Furthermore, aPC efficiently blocked oncolytic virus-induced upregulation of PD-L1 and CTLA-4. These findings indicate that oncolytic adenoviruses can reprogramme the immunosuppressive TME, while ICIs can prevent immune escape after oncolytic virus therapy by reducing the expression of immune checkpoint molecules. Our results provide a mutually reinforcing strategy for clinical combination immunotherapy.


Subject(s)
Triple Negative Breast Neoplasms , Tumor Microenvironment , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , B7-H1 Antigen , CTLA-4 Antigen , Cell Line, Tumor , Disease Models, Animal , Humans , Immunotherapy/methods , Mice , Triple Negative Breast Neoplasms/therapy
5.
Stem Cells Int ; 2021: 6662831, 2021.
Article in English | MEDLINE | ID: mdl-33747095

ABSTRACT

Paraquat (PQ) poisoning can cause acute lung injury and progress to pulmonary fibrosis and eventually death without effective therapy. Mesenchymal stem cells (MSCs) and hepatocyte growth factor (HGF) have been shown to partially reverse this damage. MSCs can be derived from bone marrow (BM-MSCs), adipose tissue (AD-MSCs), umbilical cord (UC-MSCs), dental pulp (DPSCs), and other sources. The biological characteristics of MSCs are specific to the tissue source. To develop an effective treatment for PQ poisoning, we compared the anti-inflammatory and antifibrotic effects of UC-MSCs and DPSCs and chose and modified a suitable source with HGF to investigate their therapeutic effects in vitro and in vivo. In this study, MSCs' supernatant was beneficial to the viability and proliferation of human lung epithelial cell BEAS-2B. Inflammatory and fibrosis-related cytokines were analyzed by real-time PCR. The results showed that MSCs' supernatant could suppress the expression of proinflammatory and profibrotic cytokines and increase the expression of anti-inflammatory and antifibrotic cytokines in BEAS-2B cells and human pulmonary fibroblast MRC-5. Extracellular vesicles (EVs) derived from MSCs performed more effectively than MSCs' supernatant. The effect of DPSCs was stronger than that of UC-MSCs and was further strengthened by HGF modification. PQ-poisoned mice were established, and UC-MSCs, DPSCs, and DPSCs-HGF were administered. Histopathological assessments revealed that DPSCs-HGF mitigated lung inflammation and collagen accumulation more effectively than the other treatments. DPSCs-HGF reduced lung permeability and increased the survival rate of PQ mice from 20% to 50%. Taken together, these results indicated that DPSCs can suppress inflammation and fibrosis in human lung cells better than UC-MSCs. The anti-inflammatory and antifibrotic effects were significantly enhanced by HGF modification. DPSCs-HGF ameliorated pulmonitis and pulmonary fibrosis in PQ mice, effectively improving the survival rate, which might be mediated by paracrine mechanisms. The results suggested that DPSCs-HGF transplantation was a potential therapeutic approach for PQ poisoning.

6.
Oxid Med Cell Longev ; 2020: 8032187, 2020.
Article in English | MEDLINE | ID: mdl-32855767

ABSTRACT

The metastatic potential of colorectal cancer (CRC) is intensively promoted by the tumor microenvironment (TME) in a paracrine manner. As a pleiotropic inflammatory cytokine, Interleukin-6 (IL-6) is produced and involved in CRC, the same scenario where integrin αvß6 also becomes upregulated. However, the relationship between IL-6 and integrin αvß6 as well as their involvement in the crosstalk between CRC and TME remains largely unclear. In the present study, we demonstrated a positive correlation between the expression of IL-6 and integrin ß6 in CRC samples. The mutually promotive interaction between CRC and TME was further determined by an indirect coculture system. CRC cells could augment the secretion of IL-6 from fibroblasts, which in return induced invasion and integrin ß6 expression of CRC cells. Through the classic IL-6 receptor/STAT-3 signaling pathway, IL-6 mediated the upregulation of integrin ß6, which was involved in the invasion and epithelial-mesenchymal transition of CRC cells induced by IL-6. Taken together, our results reveal a paracrine crosstalk between IL-6 signals originating from the TME and increased the integrin ß6 level of CRC. IL-6 induces CRC invasion via upregulation of integrin ß6 through the IL-6 receptor/STAT-3 signaling pathway. Combined inhibition of IL-6 along with integrin ß6-targeted strategy may indicate new directions for antitumor strategies for CRC.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Integrin beta Chains/genetics , Interleukin-6/metabolism , Up-Regulation , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Fibroblasts/metabolism , Humans , Male , Middle Aged , Neoplasm Invasiveness , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Up-Regulation/genetics
7.
Cell Biol Int ; 44(11): 2243-2252, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32716109

ABSTRACT

Conditions in space, such as microgravity, may affect the hematopoietic and bone marrow-derived mesenchymal stromal cells (BM-MSCs) of astronauts. However, to date, few detailed phenotype change data about the different type of hematopoietic cells have reported. In this study, C57BL/6 mice were randomly divided into two groups: a control group (control) and a hindlimb suspension group (treated). After four weeks of hindlimb suspension, we found that this simulated microgravity (sµg) condition could increase the percentage of monocytes and macrophages and decrease the percentage of B lymphocytes and mature red cells in bone marrow. The percentage of B lymphocytes in the spleen and the red blood cell count in peripheral blood also decreased, consistent with the response of bone marrow. The cytoskeleton in the BM-MSCs was disrupted. The expression levels of hematopoietic-related genes, such as fms-like tyrosine kinase-3 ligand, granulocyte-macrophage colony stimulating factor, interleukin-3, and adipogenic differentiation associated genes, leptin and proliferator-activated receptor γ type 2, were upregulated under sµg conditions. These results indicated that simulating microgravity can affect the phenotype of certain types of hematopoietic cells and the morphology and gene expression pattern of BM-MSCs.


Subject(s)
Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Weightlessness/adverse effects , Adipogenesis , Animals , B-Lymphocytes , Bone Marrow , Bone Marrow Cells/cytology , Cell Differentiation , Cells, Cultured , Female , Hematopoietic Stem Cells/metabolism , Hindlimb Suspension/adverse effects , Macrophages , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Monocytes , Weightlessness Simulation/methods
8.
Stem Cell Res Ther ; 11(1): 229, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32522231

ABSTRACT

BACKGROUND: To investigate the therapeutic effect of human dental pulp stem cells (DPSCs) transfected with adenovirus expressing hepatocyte growth factor (HGF) in a mouse model of collagen-induced arthritis (CIA). METHODS: DPSCs were modified with Ad-HGF to produce HGF-overexpressing DPSCs, DPSCs-HGF. In experimental mouse CIA model, DPSCs-HGF and DPSCs-Null (modified with Ad-Null) were engrafted via intravenously after disease onset, which was determined by the presence of joint swelling. The therapeutic effects on joints were evaluated at 49 days after collagen injection by histopathological analysis and microcomputed tomography imaging. The inflammatory cytokines were analyzed both in sera and joints via MILLIPLEX kit and immunohistochemical staining, respectively, and the regulatory T cells (Tregs) were analyzed in peripheral blood by using flow cytometry. Furthermore, primary fibroblast-like synoviocytes were isolated, colony formation analysis and FACS were performed to evaluate the effect of HGF on the proliferation and cell cycle of FLSs. Western blot assay was carried out to clarify the signal pathway of HGF-cMet. RESULTS: We found that without HGF modification, DPSC transfusion was helpful in controlling autoimmune status, local synovitis, and bone erosion after intravenous administration. However, HGF-modified DPSCs have dual role in rheumatoid arthritis (RA). In the early phase, HGF overexpression inhibited RA progression by its immunosuppressive effects, while in the late phase, HGF promoted synovitis by activating fibroblast-like synoviocytes to produce pathogenic IL-6, accelerating cell proliferation and inducing apoptosis resistance via phosphorylating the c-Met/Akt pathway. The overall effect of HGF modification attenuated the therapeutic effect of DPSCs. CONCLUSIONS: Our study provides a comprehensive evaluation of the therapeutic effect of DPSCs in the mouse model and a primary answer to the divergence of whether HGF is harmful or helpful in RA.


Subject(s)
Arthritis, Rheumatoid , Synoviocytes , Animals , Arthritis, Rheumatoid/therapy , Cell Proliferation , Cells, Cultured , Dental Pulp , Hepatocyte Growth Factor/genetics , Mice , Stem Cells , X-Ray Microtomography
9.
Pathol Res Pract ; 216(7): 153022, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32534716

ABSTRACT

OBJECTIVE: Integrin αvß6 is associated with an extremely aggressive cancer phenotype. However, little is known about the clinicopathological significance and prognostic value of integrin αvß6 in human hilar cholangiocarcinoma. METHODS: In the present study, bioinformatics analysis demonstrated a significant increase of integrin ß6 gene expression in cholangiocarcinoma tissues compared to non-tumorous tissues, which was further validated in clinical samples through RT-qPCR and western blotting analyses. Integrin αvß6 was observed to be expressed in 48.6% of tumors, and its expression was related to a poor tumor differentiation (p = 0.002), lymph node metastasis (p<0.001) and advanced TNM stage (p=0.001). Furthermore, patients who were αvß6-positive showed a significantly shorter overall survival period than those who were αvß6-negative (p=0.004). Multivariate analysis confirmed that integrin αvß6 was an independent prognostic factor (p=0.002). In addition, loss- and gain-of-function assays showed integrin αvß6 not only played an important role in colony formation, but also protected cholangiocarcinoma cells from cisplatin-induced growth inhibition and apoptosis. ERK/MAPK signaling pathway was involved in integrin αvß6-mediated resistance of cholangiocarcinoma cells to cisplatin. CONCLUSIONS: Taken together, the present findings revealed that integrin αvß6 could serve as a potential prognostic predictor and contribute to cisplatin resistance, which might prove to be a promising target candidate for the clinical intervention of human hilar cholangiocarcinoma.


Subject(s)
Antigens, Neoplasm/metabolism , Bile Duct Neoplasms/pathology , Biomarkers, Tumor/analysis , Integrins/metabolism , Klatskin Tumor/pathology , Aged , Antigens, Neoplasm/genetics , Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Female , Humans , Integrins/genetics , Male , Middle Aged , Prognosis
10.
Cancer Gene Ther ; 27(12): 923-933, 2020 12.
Article in English | MEDLINE | ID: mdl-32307442

ABSTRACT

LIGHT, also known as tumor-necrosis factor (TNF) superfamily member 14 (TNFSF14), is predominantly expressed on activated immune cells and some tumor cells. LIGHT is a pivotal regulator both for recruiting and activating immune cells in the tumor lesions. In this study, we armed human telomerase reverse transcriptase (TERT) promoter controlled oncolytic adenovirus with LIGHT to generate rAd.Light. rAd.Light effectively transduced both human and mouse breast tumor cell lines in vitro, and expressed LIGHT protein on the surface of tumor cells. Both rAd.Null, and rAd.Light could replicate in human breast cancer cells, and produced cytotoxicity to human and mouse mammary tumor cells. rAd.Light induced apoptosis resulting in tumor cell death. Using a subcutaneous model of 4T1 cells in BALB/c mice, rAd.Light was delivered intratumorally to evaluate the anti-tumor responses. Both rAd.Light and rAd.Null significantly inhibited the tumor growth, but rAd.Light produced much stronger anti-tumor effects. Histopathological analysis showed the infiltration of T lymphocytes in the tumor tissues. rAd.Light also induced stronger cellular apoptosis than rAd.Null in the tumors. Interestingly, on day 15, compared to rAd.Null, there was a significant reduction of Tregs following rAd.Light treatment. rAd.Light significantly increased Th1 cytokine interleukin (IL)-2 expression, and reduced Th2 cytokines expression, such as transforming growth factor ß (TGF-ß) and IL-10 in the tumors. These results suggest rAd.Light induced activation of anti-tumor immune responses. In conclusion, rAd.Light produced anti-tumor effect in a subcutaneous model of breast cancer via inducing tumor apoptosis and evoking strong anti-tumor immune responses. Therefore, rAd.Light has great promise to be developed as an effective therapeutic approach for the treatment of breast cancer.


Subject(s)
Mammary Neoplasms, Animal/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C
11.
Cell Mol Immunol ; 17(3): 261-271, 2020 03.
Article in English | MEDLINE | ID: mdl-30911117

ABSTRACT

OBJECTIVES: Anti-citrullinated protein antibodies (ACPAs) are a group of autoantibodies targeted against citrullinated proteins/peptides and are informative rheumatoid arthritis (RA) biomarkers. ACPAs also play a crucial role in RA pathogenesis, and their underlying mechanism merits investigation. METHODS: Immunohistochemical (IHC) assays were carried out to determine IL-1ß levels in ACPA+ and ACPA- RA patients. PBMC-derived monocytes were differentiated into macrophages before stimulation with ACPAs purified from RA patients. The localization and interaction of molecules were analyzed by confocal microscopy, co-IP, and surface plasmon resonance. RESULTS: In our study, we found that IL-1ß levels were elevated in ACPA+ RA patients and that ACPAs promoted IL-1ß production by PBMC-derived macrophages. ACPAs interacted with CD147 to enhance the interaction between CD147 and integrin ß1 and, in turn, activate the Akt/NF-κB signaling pathway. The nuclear localization of p65 promoted the expression of NLRP3 and pro-IL-1ß, resulting in priming. Moreover, ACPA stimulation activated pannexin channels, leading to ATP release. The accumulated ATP bound to the P2X7 receptor, leading to NLRP3 inflammasome activation. CONCLUSIONS: Our study suggests a new hypothesis regarding IL-1ß production in RA involving ACPAs, which may be a potential therapeutic target in RA treatment.


Subject(s)
Anti-Citrullinated Protein Antibodies/immunology , Arthritis, Rheumatoid/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Arthritis, Rheumatoid/pathology , Female , Humans , Macrophages/pathology , Male , THP-1 Cells
12.
Mol Med Rep ; 19(1): 213-220, 2019 01.
Article in English | MEDLINE | ID: mdl-30483783

ABSTRACT

Natural killer (NK) cells recognize stress­activated NK group 2, member D (NKG2D) ligands in tumors. In the present study, the expression levels of NKG2D ligands were examined in four lung cancer cell lines (A549, PLA801D, NCI­H157 and NCI­H520). In the A549 cells, the expression of MHC class I polypeptiderelated sequence (MIC)A/B and UL16 binding protein (ULBP)1 was weak, the expression of ULBP2 was typical, and neither ULBP3 nor ULBP4 were expressed. The mechanism underlying the regulatory effect of a cancer treatment agent on the expression of NKG2D ligands was investigated using the proteasome inhibitor MG132. Following treatment for 8 h with MG132, the transcription levels of MICB and ULBP1 were upregulated 10.62­ and 11.09­fold, respectively, and the expression levels of MICB and ULBP1 were increased by 68.18 and 23.65%, respectively. Notably, MICB exhibited significant time­dependent change. MG132 increased the transcription of MICB by acting at a site in the 480­bp MICB upstream promoter. The activity of the MICB promoter was upregulated 1.77­fold following treatment with MG132. MG132 treatment improved the cytotoxicity of NK cells, which was partially blocked by an antibody targeting NKG2D, and more specifically the MICB molecule. The expression of MICB induced by MG132 was inhibited by KU­55933 [ataxia telangiectasia mutated (ATM) kinase inhibitor], wortmannin (phosphoinositide 3 kinase inhibitor) and caffeine (ATM/ATM­Rad3­related inhibitor). The phosphorylation of checkpoint kinase 2 (Chk2), an event associated with DNA damage, was observed following treatment with MG132. These results indicated that MG132 selectively upregulates the expression of MICB in A549 cells, and increases the NKG2D­mediated cytotoxicity of NK cells. The regulatory effect of MG132 may be associated with the activation of Chk2, an event associated with DNA damage. The combination of MG132 with NK cell immunotherapy may have a synergistic effect that improves the therapeutic effect of lung cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/metabolism , Leupeptins/pharmacology , Lung Neoplasms/metabolism , A549 Cells , DNA Damage , Histocompatibility Antigens Class I/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Promoter Regions, Genetic , Transcriptional Activation
13.
J Immunol Res ; 2018: 1687097, 2018.
Article in English | MEDLINE | ID: mdl-30155491

ABSTRACT

miR-362 is a recently discovered member of the microRNA family, and it modulates a variety of physical activities and plays an important role in the occurrence and development of many tumors. However, the biological functions of hsa-miR-362-5p in non-small-cell lung carcinoma (NSCLC) are unknown. Transwell assay and colony formation were used to determine the migration, invasion, and proliferation of NSCLC cells in vitro. A subcutaneous tumor model in nude mice was established to detect NSCLC tumor growth in vivo. The direct binding of miR-362 to the 3'UTR of Semaphorin 3A (Sema3A) was confirmed by luciferase reporter assay. In this study, we found that the level of miR-362 was higher in NSCLC tissues than in adjacent normal tissues and that the level of miR-362 expression was also elevated in five NSCLC cell lines (A549, 95-D, H1299, H292, and H460) relative to a human normal lung epithelial cell line (BEAS2B). Furthermore, miR-362 promoted NSCLC cell invasion, migration, and colony formation in vitro and tumor formation in vivo. Next, we identified the miR-362 target gene Sema3A, which is significantly correlated with metastasis. Sema3A expression was increased in normal tissues relative to NSCLC tissues. This result is consistent with the fact that miR-362 expression is negatively correlated with Sema3A expression in clinical tissue samples and indicated that miR-362 can regulate Sema3A expression in NSCLC cells and consequently affect NSCLC invasion, migration, and colony formation. Taken together, these findings on the newly identified miR-362/Sema3A axis elucidate the molecular mechanism of NSCLC invasion and migration and could lead to a potential therapeutic target in NSCLC treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Neoplasms, Experimental/genetics , Semaphorin-3A/genetics , Animals , Cell Movement , Cell Proliferation , Down-Regulation , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Semaphorin-3A/metabolism
14.
Autoimmun Rev ; 17(9): 845-853, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30007856

ABSTRACT

The production of anti-citrullinated peptide antibodies (ACPAs) requires the participation of both innate immunity and adaptive immunity. On the one hand, activated innate immunity is able to produce citrullinated auto-antigens that fuel autoimmunity and provide an inflammatory environment that facilitates the breach of self-tolerance, proliferation of self-reactive T/B cells and the production of ACPAs. On the other hand, after their production by plasma B cells, ACPAs are also able to interact with innate immunity to exacerbate the manifestation and chronicity of rheumatoid arthritis (RA). This article discusses the roles of citrullinated peptides and ACPA played in innate immunity and autoimmunity. In addition, we emphasise the relationships between environmental factors and innate immunity, as well as the pathogenic function of ACPAs per se. In doing so, we hope to provide fundamental knowledge of RA pathogenesis and reveal potential therapeutic targets in RA treatment.


Subject(s)
Adaptive Immunity/immunology , Anti-Citrullinated Protein Antibodies/therapeutic use , Arthritis, Rheumatoid/drug therapy , Immunity, Innate/immunology , Anti-Citrullinated Protein Antibodies/pharmacology , Arthritis, Rheumatoid/immunology , Humans
15.
Medicine (Baltimore) ; 97(16): e0343, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29668584

ABSTRACT

The aim of this study was to determine whether combined utilization of untimed single urine monocyte chemoattractant protein 1 (uMCP-1) and tumor necrosis factor (TNF)-like weak inducer of apoptosis (uTWEAK) could serve as a screening test for proteinuria in patients with lupus nephritis (LN).A case-control study that contained 39 biopsy-proven LN patients, 20 non-LN systemic lupus erythematosus (SLE) patients, and 10 healthy controls (HCs) were carried out. Correlations between uMCP-1, uTWEAK, and traditional clinical markers were analyzed by Spearman correlation test. Diagnostic values of uMCP-1, uTWEAK, and urine albumin/creatinine ratio (uACR) in the assessment of proteinuria were investigated by receiver operating characteristic (ROC) curves.Biopsy-proven LN patients showed higher levels of uMCP-1 and uTWEAK than non-LN patients. uMCP-1 and uTWEAK were elevated in renal active patients (rSLEDAI ≥4). Both uMCP-1 and uTWEAK showed significant correlation with patients' rSLEDAI, 24-hour urine proteinuria (24hr UP), and anti-double-stranded DNA (anti-dsDNA) antibodies. No correlations of these 2 biomarkers between cystatin C (Cys-C), creatinine (Cr), and blood urea nitrogen (BUN) were observed. An algorithm combining the moderate sensitivity of uMCP-1 and high specificity of uTWEAK displayed great specificity and sensitivity for proteinuria screening.Both uMCP-1 and uTWEAK were positively correlated with the impairments of LN, and the combined utility of untimed single uMCP-1 and uTWEAK might be used as potential predictors for proteinuria in LN.


Subject(s)
Chemokine CCL2/urine , Cytokine TWEAK/urine , Lupus Nephritis , Proteinuria , Urinalysis , Adult , Biomarkers/urine , Biopsy/methods , Creatinine/analysis , Female , Humans , Kidney/pathology , Lupus Nephritis/complications , Lupus Nephritis/diagnosis , Male , Mass Screening/methods , Mass Screening/statistics & numerical data , Predictive Value of Tests , Proteinuria/diagnosis , Proteinuria/etiology , ROC Curve , Sensitivity and Specificity , Statistics as Topic , Urinalysis/methods , Urinalysis/statistics & numerical data
16.
Sci Rep ; 8(1): 427, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323140

ABSTRACT

Mesenchymal stem cells (MSCs) are multi-potent cells that are self-renewable and possess the potential to differentiate into multiple lineages. Several studies demonstrated that MSCs could regulate a Th17/Treg balance and could be a potential therapeutic target for Rheumatoid Arthritis (RA). A20 is highly expressed in many cell types after the stimulation of TNF-α, where it may inhibit pro-inflammatory cytokine secretion. However, the expression of A20 in BM-MSCs in RA is not fully understood. In our study, we found that A20 was decreased in RA patients' bone marrow MSCs (BM-MSCs), and with more IL-6 secretion, the balance of Th17/Treg was broken. In CIA mice, we found a moderate A20 decrease in mice MSCs as compared with those of control group in mRNA and protein levels. However, the IL-6 expression was increased. After umbilical cord MSCs treatment, A20 and IL-6 expressions were equal to the control group. Thus, our study indicates that loss of A20 in MSCs regulates the Th17/Treg balance in RA and the regulatory role of A20 in pro-inflammatory IL-6 production could be a potential target for the transfer of MSCs in RA adoptive therapy.


Subject(s)
Arthritis, Rheumatoid/metabolism , Down-Regulation , Mesenchymal Stem Cells/cytology , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Adult , Aged , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/therapy , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Female , Humans , Interleukin-6/metabolism , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Middle Aged , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics
17.
Int J Mol Sci ; 19(2)2018 Jan 23.
Article in English | MEDLINE | ID: mdl-29360760

ABSTRACT

The maintenance of ordinal cell cycle phases is a critical biological process in cancer genesis, which is a crucial target for anti-cancer drugs. As an important natural isoquinoline alkaloid from Chinese herbal medicine, Berberine (BBR) has been reported to possess anti-cancer potentiality to induce cell cycle arrest in hepatocellular carcinoma cells (HCC). However, the underlying mechanism remains to be elucidated. In our present study, G0/G1 phase cell cycle arrest was observed in berberine-treated Huh-7 and HepG2 cells. Mechanically, we observed that BBR could deactivate the Akt pathway, which consequently suppressed the S-phase kinase-associated protein 2 (Skp2) expression and enhanced the expression and translocation of Forkhead box O3a (FoxO3a) into nucleus. The translocated FoxO3a on one hand could directly promote the transcription of cyclin-dependent kinase inhibitors (CDKIs) p21Cip1 and p27Kip1, on the other hand, it could repress Skp2 expression, both of which lead to up-regulation of p21Cip1 and p27Kip1, causing G0/G1 phase cell cycle arrest in HCC. In conclusion, BBR promotes the expression of CDKIs p21Cip1 and p27Kip1 via regulating the Akt/FoxO3a/Skp2 axis and further induces HCC G0/G1 phase cell cycle arrest. This research uncovered a new mechanism of an anti-cancer effect of BBR.


Subject(s)
Berberine/pharmacology , Cell Cycle Checkpoints/drug effects , Forkhead Box Protein O3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Protein Transport , S-Phase Kinase-Associated Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...