Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
J Cell Mol Med ; 28(10): e18395, 2024 May.
Article in English | MEDLINE | ID: mdl-38774995

ABSTRACT

Tumour-associated macrophages (TAMs), encompassing M1 and M2 subtypes, exert significant effects on osteosarcoma (OS) progression and immunosuppression. However, the impacts of TAM-derived biomarkers on the progression of OS remains limited. The GSE162454 profile was subjected to single-cell RNA (scRNA) sequencing analysis to identify crucial mediators between TAMs and OS cells. The clinical features, effects and mechanisms of these mediators on OS cells and tumour microenvironment were evaluated via biological function experiments and molecular biology experiments. Phosphodiesterase 4C (PDE4C) was identified as a pivotal mediator in the communication between M2 macrophages and OS cells. Elevated levels of PDE4C were detected in OS tissues, concomitant with M2 macrophage level, unfavourable prognosis and metastasis. The expression of PDE4C was observed to increase during the conversion process of THP-1 cells to M2 macrophages, which transferred the PDE4C mRNA to OS cells through exosome approach. PDE4C increased OS cell proliferation and mobility via upregulating the expression of collagens. Furthermore, a positive correlation was observed between elevated levels of PDE4C and increased TIDE score, decreased response rate following immune checkpoint therapy, reduced TMB and diminished PDL1 expression. Collectively, PDE4C derived from M2 macrophages has the potential to enhance the proliferation and mobility of OS cells by augmenting collagen expression. PDE4C may serve as a valuable biomarker for prognosticating patient outcomes and response rates following immunotherapy.


Subject(s)
Bone Neoplasms , Cyclic Nucleotide Phosphodiesterases, Type 4 , Immunotherapy , Macrophages , Osteosarcoma , Tumor Microenvironment , Humans , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Gene Expression Regulation, Neoplastic , Immunotherapy/methods , Macrophages/metabolism , Macrophages/immunology , Neoplasm Metastasis , Osteosarcoma/pathology , Osteosarcoma/immunology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/therapy , Prognosis , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
2.
Artif Cells Nanomed Biotechnol ; 52(1): 156-174, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38423139

ABSTRACT

Osteoarthritis (OA) is a degenerative disease closely associated with Anoikis. The objective of this work was to discover novel transcriptome-based anoikis-related biomarkers and pathways for OA progression.The microarray datasets GSE114007 and GSE89408 were downloaded using the Gene Expression Omnibus (GEO) database. A collection of genes linked to anoikis has been collected from the GeneCards database. The intersection genes of the differential anoikis-related genes (DEARGs) were identified using a Venn diagram. Infiltration analyses were used to identify and study the differentially expressed genes (DEGs). Anoikis clustering was used to identify the DEGs. By using gene clustering, two OA subgroups were formed using the DEGs. GSE152805 was used to analyse OA cartilage on a single cell level. 10 DEARGs were identified by lasso analysis, and two Anoikis subtypes were constructed. MEgreen module was found in disease WGCNA analysis, and MEturquoise module was most significant in gene clusters WGCNA. The XGB, SVM, RF, and GLM models identified five hub genes (CDH2, SHCBP1, SCG2, C10orf10, P FKFB3), and the diagnostic model built using these five genes performed well in the training and validation cohorts. analysing single-cell RNA sequencing data from GSE152805, including 25,852 cells of 6 OA cartilage.


Subject(s)
Anoikis , Osteoarthritis , Humans , Anoikis/genetics , Machine Learning , Cadherins , Osteoarthritis/diagnosis , Osteoarthritis/genetics , Sequence Analysis, RNA , Shc Signaling Adaptor Proteins
3.
Eur J Pharm Sci ; 194: 106706, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38244809

ABSTRACT

Cervical cancer is the leading cause of death among gynecological malignant tumors, especially due to the poor prognosis of patients with advanced tumors due to recurrence, metastasis, and chemotherapy resistance. Therefore, exploring new antineoplastic drugs with high efficacy and low toxicity may bring new expectations in patients with cervical cancer. Natural products and their derivatives exert an antitumor activity. Therefore, in this work, combined with network pharmacology analysis and experimental validation, we investigated the anti-cervical cancer activity and molecular mechanism of a new trifluoromethyl quinoline (FKL) derivative in vivo and in vitro. FKL117 inhibited the proliferation of cervical cancer cells in a dose and time-dependent manner, induced apoptosis in HeLa cells, arrested the cell cycle in the G2/M phase, and regulated the expression of the apoptotic and cell cycle-related proteins Bcl-2, Bax, cyclin B1, and CDC2. We used online databases to obtain HDAC1 as one of the possible targets of FKL117 and the target binding and binding affinity were modeled by molecular docking. The results showed that FKL117 formed a hydrogen bond with HDAC1 and had good binding ability. We found that FKL117 targeted to inhibit the expression and function of HDAC1 and increased the acetylation of histone H3 and H4, which was also confirmed in vivo. The migration of HMGB1 from the nucleus to the cytoplasm further verified the above results. In conclusion, our study suggested that FKL117 might be used as a novel candidate for targeting the inhibition of HDAC1 against cervical cancer.


Subject(s)
Quinolines , Uterine Cervical Neoplasms , Female , Humans , Histones/metabolism , Uterine Cervical Neoplasms/drug therapy , HeLa Cells , Acetylation , Molecular Docking Simulation , Cell Line, Tumor , Apoptosis , Quinolines/pharmacology , Quinolines/therapeutic use , Cell Proliferation , Histone Deacetylase 1/metabolism
4.
J Int Adv Otol ; 19(6): 523-528, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38088327

ABSTRACT

BACKGROUND: We aimed to analyze the independent risk factors that affect the treatment outcomes of residual symptoms of cured benign paroxysmal positional vertigoand to construct a nomogram model. METHODS: A total of 186 benign paroxysmal positional vertigo patients who were treated in our hospital from June 2019 to August 2021 were selected. According to whether there were residual symptoms, they were divided into a group with residual symptoms (n=82) and a group without residual symptoms (n = 104). The logistic regression model was used to analyze the independent risk factors affecting the treatment outcomes, and the results were incorporated into R software to establish a nomogram model for verification. RESULTS: The incidence rate of residual symptoms in the 186 patients was 44.09% (82/186). Logistic regression analysis showed that age, course of disease, number of maneuvers, anxiety state, diabetes mellitus, and hypertension were independent risk factors affecting the treatment outcomes of residual symptoms after cured benign paroxysmal positional vertigo. The area under the receiver operating characteristic curve of the nomogram model was 0.938. The calibration curve was fitted well (χ2 = 8.165, P = .417). CONCLUSION: The nomogram model constructed based on age, course of disease, number of maneuvers, anxiety state, diabetes mellitus, and hypertension had a high predictive value for the treatment outcomes of residual symptoms in benign paroxysmal positional vertigo patients.


Subject(s)
Diabetes Mellitus , Hypertension , Humans , Benign Paroxysmal Positional Vertigo/diagnosis , Benign Paroxysmal Positional Vertigo/epidemiology , Benign Paroxysmal Positional Vertigo/therapy , Nomograms , Risk Factors , Patient Positioning
5.
J Cancer ; 14(11): 2066-2074, 2023.
Article in English | MEDLINE | ID: mdl-37497413

ABSTRACT

Epidemiological data have shown a positive correlation between lipid levels and tumor occurrence, such as the correlation between tumor frequency and aggressiveness, and cardiovascular disease, obesity, type 2 diabetes mellitus, and hyperinsulinemia. Therefore, reducing fat accumulation or weakening lipid metabolism may affect the carcinogenic processes of cells. Many studies have shown that traditional Chinese Medicine (TCM) has obvious advantages over traditional therapies in terms of fewer side effects, lower toxicity, and lower economic burden. This paper reviews the mechanism by which TCM regulates lipid metabolism and its antitumor effect through this regulation, with the aim of elucidating the bioactive compounds in TCM with good efficacy and few side effects that can provide promising therapeutic drugs for targeting lipid metabolism reprogramming in cancer.

6.
Cell Commun Signal ; 21(1): 46, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36864422

ABSTRACT

BACKGROUND: Early metastasis is a hallmark of osteosarcoma (OS), a highly common type of malignant tumor. Members of the potassium inwardly rectifying channel family exert oncogenic effects in various cancers. However, the role of the potassium inwardly rectifying channel subfamily J member 2 (KCNJ2) in OS is unclear. METHODS: The expression of KCNJ2 in OS tissues and cell lines was measured using bioinformatic analysis, immunohistochemistry, and western blotting. Wound-healing assays, Transwell assays, and lung metastasis models were used to analyze the effects of KCNJ2 on mobility of OS cells. The molecular mechanisms linking KCNJ2 and HIF1α in OS were explored by mass spectrometry analysis, immunoprecipitation, ubiquitination detection, and chromatin-immunoprecipitation quantitative real-time polymerase chain reaction. RESULTS: KCNJ2 was found to be overexpressed in advanced-stage OS tissues, as well as in cells with high metastatic potential. High expression of KCNJ2 was associated with a shorter survival rate of OS patients. KCNJ2-inhibition repressed the metastasis of OS cells, whereas KCNJ2-elevation induced the opposite effects. Mechanistically, KCNJ2 binds to HIF1α and inhibits its ubiquitination, thus increasing the expression of HIF1α. Interestingly, HIF1α binds directly to the KCNJ2 promoter and increases its transcription under hypoxic conditions. CONCLUSION: Taken together, our results indicated that a KCNJ2/HIF1α positive feedback loop exists in OS tissues, which significantly promotes OS cell metastasis. This evidence may contribute to the diagnosis and treatment of OS. Video Abstract.


Subject(s)
Bone Neoplasms , Osteosarcoma , Potassium Channels, Inwardly Rectifying , Humans , Feedback , Biological Assay , Cell Line , Bone Neoplasms/genetics , Potassium Channels, Inwardly Rectifying/genetics
7.
Pharmacology ; 108(3): 224-237, 2023.
Article in English | MEDLINE | ID: mdl-36889301

ABSTRACT

BACKGROUND: Autophagy is a lysosome-mediated catabolic process that maintains cell homeostasis and survival. It occurs not only in normal cells such as cardiac muscle cells, neurons, and pancreatic acinar cells but also in various benign and malignant tumors. The abnormal level of intracellular autophagy is closely related to multiple pathophysiological processes, including aging, neurodegeneration, infectious diseases, immune disorders, and cancer. Autophagy mainly plays a dual role in life and death by regulating cell survival, proliferation, and death, thus being involved in the occurrence, development, and treatment of cancer. It is also involved in chemotherapy resistance by a dual role, since it not only promotes the occurrence of drug resistance but also reverses it. Previous findings suggest that the regulation of autophagy can be used as an effective strategy in tumor therapy. SUMMARY: Recent studies found that small molecules from natural products and their derivatives exert anticancer activity by regulating the level of autophagy in tumor cells. KEY MESSAGES: Therefore, this review article describes the mechanism of autophagy, the role of autophagy in normal cells and tumor cells, and the research progress on the anticancer molecular mechanism of targets regulating cell autophagy. The aim is to provide a theoretical basis for developing autophagy inhibitors or activators to improve anticancer efficacy.


Subject(s)
Neoplasms , Humans , Neoplasms/drug therapy , Lysosomes , Autophagy , Signal Transduction/physiology , Cell Survival
8.
Biomark Med ; 15(3): 191-200, 2021 02.
Article in English | MEDLINE | ID: mdl-33496611

ABSTRACT

Background: Endothelial dysfunction is considered to be involved in the pathogenesis of cerebral small vessel disease (CSVD). Endothelial progenitor cells are associated with endothelial dysfunction. The present study was designed to investigate the correlation between the populations of circulating CD34-positive cells and endothelial progenitor cells and CSVD burden. Methodology & results: A total of 364 patients with confirmed diagnosis of CSVD were included in this prospective study. Multiple ordinal logistic regression analyses showed that subjects with higher CSVD burden had significantly decreased circulating CD34+ cell level (odds ratio [OR], 0.42; p = 0.034) and significantly increased levels of circulating CD34+CD133+CD309+ and CD34+CD133+ cells (OR 1.07, p = 0.031; OR 1.03, p = 0.001, respectively), compared with patients with lower CSVD burden. Conclusion: The findings suggest that the levels of circulating CD34+ cells, CD34+CD133+CD309+ cells and CD34+CD133+ cells may be used as potential biomarkers to monitor the disease progression of CSVD.


Subject(s)
Cerebral Small Vessel Diseases , Adult , Aged , Antigens, CD34 , Endothelial Progenitor Cells , Humans , Male , Middle Aged , Prospective Studies , Stem Cells , Vascular Endothelial Growth Factor Receptor-2
9.
Clin Pharmacol Ther ; 110(1): 229-237, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33483974

ABSTRACT

Overexpression of AKR1C3, an aldo-keto reductase, was recently discovered in liver cancers. In this study, an inverse correlation between AKR1C3 expression and survival of patients with liver cancer was observed. AKR1C3 inhibitors, however, failed to suppress liver cancer cell growth. The prodrug TH3424, which releases a DNA alkylating reagent upon reduction by AKR1C3, was developed to target tumors with overexpression of AKR1C3. TH3424 showed specific killing of liver cancer cells with AKR1C3 overexpression both in vitro and in vivo. In patient-derived mouse xenograft models, TH3424 at doses as low as 1.5 mg/kg eliminated liver tumors with no apparent toxicity. Therefore, TH3424 is a promising drug candidate for liver cancer and other types of cancers overexpressing AKR1C3.

10.
Leuk Lymphoma ; 61(7): 1660-1668, 2020 07.
Article in English | MEDLINE | ID: mdl-32091283

ABSTRACT

AKR1C3 overexpression has been reported in various types of cancers, including T-ALL. AST-006 (TH-3424), an AKR1C3-specific prodrug, was reported recently to have potent cytotoxicity against liver cancer cells overexpressing AKR1C3 and T-ALL. In this study, AST-006 demonstrated potent anti-tumor activity against different T-ALL cell lines in vitro and in vivo, including patient-derived xenograft (PDX) model. AST-006 also exhibited minimal cytotoxicity against primary human T-cells in vitro and lymphocytes in cynomolgus monkeys in vivo, indicating that AST-006 is a promising therapeutic for T-ALL.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Prodrugs , Aldo-Keto Reductase Family 1 Member C3 , Antineoplastic Agents, Alkylating , Cell Line, Tumor , Humans , Prodrugs/pharmacology , Xenograft Model Antitumor Assays
11.
Langmuir ; 35(12): 4342-4352, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30810322

ABSTRACT

In this article, 3-(3,5-di- tert-butyl-4-hydroxyphenyl) propionic acid (DBHP)-functionalized ZnO (DBHP-ZnO) nanoparticles were synthesized by decomposing the organometallic precursor Zn(DBHP)2 under alkaline conditions. This in situ surface modification method can induce small-sized ZnO nanoparticles (5 nm) and form strong linkage between DBHP and ZnO nanoparticles. DBHP as an organic compound hindered phenol antioxidant that not only improved the dispersion stability of the prepared DBHP-ZnO nanoparticles in the lubrication oil but also scavenged free radicals produced during the oxidation process of oil. Compared with DBHP, the thermal stability of the prepared composite antioxidant was greatly enhanced by introducing inorganic ZnO nanoparticles, which was proved by the results of the thermogravimetric analysis test. A rotary oxygen bomb test, pressurized differential scanning calorimetry, and free-radical-scavenging method all showed that DBHP-ZnO nanoparticles had better antioxidant properties than DBHP under high temperature in the base oil of di- iso-octylsebacate (DIOS). The activation energy of the oxidation process was used to analyze this result by the model-free methods, including the Flynn-Wall-Ozawa method and the Kissinger equation. The calculated results showed that DIOS containing DBHP-ZnO nanoparticles have the lowest reaction constant and the longest half-life period compared to those of individual DBHP and ZnO nanoparticles, which is attributed to the combined action of the organic-inorganic composites. Besides, DBHP-ZnO nanoparticles as the additive are able to improve the antiwear ability of DIOS to some extent. Therefore, the as-prepared DBHP-ZnO nanoparticles with desired dispersibility as well as better thermal stability and antioxidant ability than DBHP in the DIOS base oil could be a potential high-performance nanocomposite additive for a synthetic lubricant base oil like DIOS.

12.
Oncol Lett ; 16(1): 1259-1266, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29963199

ABSTRACT

Bispecific antibodies have been actively studied for cancer therapy due to their potent cytotoxicity against tumor cells. A number of bispecific antibody formats have exhibited strong tumor cytotoxicity in vitro and in vivo. However, effective production of bispecific antibodies remains challenging for the majority of bispecific antibody formats. In the present study, a bispecific antibody was designed that links a conventional antigen-binding fragment (Fab) against cluster of differentiation 3 antigen (CD3) to a camel single domain antibody (VHH) against human epidermal growth factor receptor 2 (HER2). This bispecific antibody may be secreted and purified efficiently from Escherichia coli culture medium. The purified bispecific antibody is able to trigger T cell-mediated HER2-specific cytotoxicity in vitro and in vivo. The data gathered in the present study suggest that this bispecific format may be applied to other tumor antigens to produce bispecific antibodies more efficiently.

13.
Oncotarget ; 9(33): 23102-23113, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29796175

ABSTRACT

Transforming growth factor beta (TGF-ß) promotes cancer growth in late stage cancers. To inhibit the TGF-ß pathway, we investigated a tumor-targeting TGF-ß receptor blocker, TTB, and its role in tumor progress. The targeted TTB comprised of the extracellular domain of the TGF-ß receptor II, the endoglin domain of TGF-ß receptor III, and the human immuno-globin IgG1 constant fragment (Fc). To enhance tumor microenvironment targeting, a RGD peptide was fused at the N-terminal of TTB. The targeted TTB exhibited potent TGF-ß neutralization activities, and inhibited cancer cell migration and invasion as well as colony formation. In xenograft models, the TTB had potent tumor inhibition activities. The TTB also attenuated the TGF-ß1-induced Smad2 phosphorylation and epithelial to mesenchymal transformation (EMT), and suppressed breast cancer metastasis. Thus, the TTB is an effective TGF-ß blocker with a potential for blocking excessive TGF-ß induced pathogenesis in vivo.

14.
PLoS One ; 13(1): e0191024, 2018.
Article in English | MEDLINE | ID: mdl-29357376

ABSTRACT

Muc1 is one of the most studied tumor antigens. However, antibodies or antibody-toxin conjugates against Muc1 have not shown significant efficacy for tumors with Muc1 overexpression. In this study, we employed bispecific antibody approach to target Muc1 positive tumor cells. A novel bispecific antibody, Muc1-Bi-1, was constructed by linking single domain antibodies, anti-Muc1-VHH and anti-CD16-VHH. Muc1-Bi-2, the humanized form of Muc1-Bi-1, was also constructed by grafting. Both Muc1-Bi bispecific antibodies can be efficiently expressed and purified from bacteria. In vitro, the Muc1-Bi bispecific antibodies can recruit Natural Killer (NK) cells to drive potent and specific cell killing of Muc1-overexpressing tumor cells. In xenograft model, the Muc1-Bi bispecific antibodies can suppress tumor growth in the presence of human peripheral blood mononuclear cells (PBMC). These data suggested that the single domain based Muc1-Bi may provide a valid strategy for targeting tumors with Muc1 overexpression.


Subject(s)
Antibodies, Bispecific/immunology , Mucin-1/immunology , Animals , Antibodies, Bispecific/genetics , CHO Cells , Cell Line, Tumor , Cell Proliferation , Cricetulus , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology
15.
Appl Immunohistochem Mol Morphol ; 26(10): 727-733, 2018.
Article in English | MEDLINE | ID: mdl-28362712

ABSTRACT

Previous studies have revealed that the activation of the epithelial-mesenchymal transition (EMT) endows metastatic properties upon cancer cells to promote invasion and migration. In this study, immunohistochemical analysis was performed in 50 cases of clear cell renal cell carcinoma (RCC) and paired normal kidney tissues. We detected the expression of vascular endothelial growth inhibitor (VEGI) and EMT markers (E-cadherin, fibronectin, and Slug) and recorded the clinical, pathologic, and follow-up (median follow-up: 79.0 mo) information. The expression of VEGI and E-cadherin was significantly lower in RCC tissues compared with normal kidney tissues (P<0.001). However, the expression of fibronectin and Slug was higher in RCC tissues (P<0.05). VEGI and EMT marker expression marginally differed in tumor size and stage. Significant differences were found in the pathologic grade (P<0.05). The Spearman correlation analysis suggested a positive correlation between VEGI and E-cadherin (r=0.451, P<0.01). A negative correlation was shown between VEGI and fibronectin (r=-0.465, P<0.01). There was also a negative correlation between VEGI and Slug (r=-0.758, P<0.01). During the 79.0 months (range, 7 to 119 mo) of follow-up, 6 patients died due to RCC, and the tumor-free survival rate was 88% (44/50). We did not find a significant correlation between VEGI/EMT markers (E-cadherin, fibronectin, and Slug) and overall survival (P>0.05). Our findings indicate that VEGI plays an important role in EMT in RCC. It suggests that VEGI may be investigated as a disease biomarker and therapeutic target in RCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell , Epithelial-Mesenchymal Transition , Kidney Neoplasms , Neoplasm Proteins/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Adult , Aged , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Disease-Free Survival , Female , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Middle Aged , Survival Rate
16.
J Immunother ; 41(1): 1-8, 2018 01.
Article in English | MEDLINE | ID: mdl-29232309

ABSTRACT

Human epidermal growth factor receptor 2 (HER2) is frequently overexpressed and activated in metastatic breast cancers. Monoclonal antibodies targeting Her2, such as trastuzumab and pertuzumab, have become important targeted therapies for patients with HER2-positive breast cancer. Both trastuzumab and pertuzumab can reduce Her2 positive tumor burden by inhibiting Her2 signaling and inducing ADCC activities (antibody dependent cell-mediated cytotoxicity). In this study, we have generated a bispecific antibody, Her2(Per)-S-Fab, by linking the pertuzumab Fab to an anti-CD16 single domain antibody. The Her2(Per)-S-Fab can be expressed and purified efficiently from Escherichia coli. In vitro and in vivo experiments showed Her2(Per)-S-Fab had potent cytotoxicity against Her2-positive tumor cells. Thus, Her2(Per)-S-Fab may provide an alternative to treat Her2-positive cancer patients.


Subject(s)
Antibodies, Bispecific/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/therapy , Immunotherapy/methods , Receptor, ErbB-2/genetics , Animals , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/genetics , Antibody-Dependent Cell Cytotoxicity , Breast Neoplasms/immunology , Escherichia coli/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mice, SCID , Molecular Targeted Therapy , Neoplasm Metastasis , Protein Engineering , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptors, IgG/immunology , Signal Transduction , Single-Domain Antibodies/genetics , Single-Domain Antibodies/therapeutic use , Trastuzumab/therapeutic use , Tumor Burden , Xenograft Model Antitumor Assays
17.
Transl Oncol ; 10(5): 780-785, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28797938

ABSTRACT

Among different cancer immunotherapy approaches, bispecific antibodies (BsAbs) are of great interest due to their ability to recruit immune cells to kill tumor cells directly. Various BsAbs against Her2 tumor cells have been proposed with potent cytotoxic activities. However, most of these formats require extensive processing to obtain heterodimeric bispecific antibodies. In this study, we describe a bispecific antibody, BiHC (bispecific Her2-CD3 antibody), constructed with a single-domain anti-Her2 and a single-chain Fv (variable fragment) of anti-CD3 in an IgG-like format. In contrast to most IgG-like BsAbs, the two arms in BiHC have different molecular weights, making it easier to separate hetero- or homodimers. BiHC can be expressed in Escherichia coli and purified via Protein A affinity chromatography. The purified BiHC can recruit T cells and induce specific cytotoxicity of Her2-expressing tumor cells in vitro. The BiHC can also efficiently inhibit the tumor growth in vivo. Thus, BiHC is a promising candidate for the treatment of Her2-positive cancers.

18.
Int J Mol Med ; 40(2): 569-575, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28656288

ABSTRACT

The present study was carried out to investigate the effects of vascular endothelial growth inhibitor 174 (VEGI174) and its functional domains (V7 and V8) on epithelial­mesenchymal transition (EMT) in renal cell carcinoma (RCC) cells in vitro. The RCC cell lines A498 and 786­O were used in this study. Based on our preliminary study, we selected full­length VEGI174 and its functional domains (V7 and V8) as the target genes in this study. Plasmids containing VEGI174, V7 or V8 transgenes were constructed and transfected into A498 and 786­O cell lines. Cytological activity was tested during cell culture. Quantitative PCR and western blot analysis were performed to determine the expression levels of EMT markers (E­cadherin, vimentin, ß­catenin and Slug). Overexpression of VEGI174, V7 or V8 did not have a significant influence on cell viability (P>0.05). The mRNA level of E­cadherin was significantly upregulated, while that of vimentin was downregulated in A498VEGIexp, A498V7exp, A498V8exp, 786­OVEGIexp, 786­OV7exp and 786­OV8exp cells compared with the cells containing the empty plasmid controls (P<0.05). The western blot results showed that changes in protein expression levels were consistent with the changes in mRNA expression. Both the mRNA and protein expression levels of ß­catenin and Slug were downregulated in the A498VEGIexp, A498V7exp, A498V8exp, 786­OVEGIexp, 786­OV7exp and 786­OV8exp cells. In conclusion, overexpression of VEGI174, V7 or V8 inhibited EMT in A498 and 786­O cells. Notably, V7 and V8 are two effective functional domains of VEGI174 that have the potential to be studied for peptide synthesis and the treatment of RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Epithelial-Mesenchymal Transition , Kidney Neoplasms/pathology , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Up-Regulation
19.
Ann Clin Lab Sci ; 47(1): 40-46, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28249915

ABSTRACT

OBJECTIVE: To establish epithelial-mesenchymal transition (EMT) models in renal cell carcinoma (RCC) cell lines. MATERIALS AND METHODS: The RCC cell lines A498 and 786-O were used in the experiment and CoCl2 was used to simulate hypoxia. Cells were cultured with different concentrations of CoCl2. Morphology and changes in cytoactivity were observed. After CoCl2 treatment, the expression of HIF-1α and the changes of EMT-related molecules (E-cadherin, fibronectin) were detected. RESULTS: Cell conjunctions of CoCl2-treated groups were loose and scattered compared to the control. CoCl2 did not promote or attenuate the viability of A498 cells at low dosage, but when the concentration of CoCl2 reached 250 µM, cell activity gradually declined. In contrast, CoCl2 induced 786-O cell proliferation in the range of 50 µ M-200 µ M, but inhibited cell growth at dosages higher than 200 µM. The expression of E-cadherin was significantly down-regulated, and fibronectin was up-regulated in both A498 and 786-O cell lines under CoCl2-simulated hypoxia in comparison with normoxic conditions (P<0.01). CONCLUSIONS: CoCl2-induced hypoxia could induce EMT in RCC cell lines. The models will help us further study the mechanisms of EMT and investigate novel therapeutic targets to inhibit tumor invasion and metastasis.


Subject(s)
Carcinoma, Renal Cell/pathology , Cobalt/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Kidney Neoplasms/pathology , Carcinoma, Renal Cell/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/metabolism
20.
Transl Res ; 180: 91-102.e1, 2017 02.
Article in English | MEDLINE | ID: mdl-27639592

ABSTRACT

Heart failure (HF) is associated with myocardial energy metabolic abnormality. Receptor-interacting protein 140 (RIP140) is an important transcriptional cofactor for maintaining energy balance in high-oxygen consumption tissues. However, the role of RIP140 in the pathologic processes of HF remains to be elucidated. In this study, we investigated the role of RIP140 in mitochondrial and cardiac functions in rodent hearts under myocardial infarction (MI) stress. MI was created by a permanent ligation of left anterior descending coronary artery and exogenous expression of RIP140 by adenovirus (Ad) vector delivery. Four weeks after MI or Ad-RIP140 treatment, cardiac function was assessed by echocardiographic and hemodynamics analyses, and the mitochondrial function was determined by mitochondrial genes expression, biogenesis, and respiration rates. In Ad-RIP140 or MI group, a subset of metabolic genes changed, accompanied with slight reductions in mitochondrial biogenesis and respiration rates but no change in adenosine triphosphate (ATP) content. Cardiac malfunction was compensated. However, under MI stress, rats overexpressing RIP140 exhibited greater repressions in mitochondrial genes, state 3 respiration rates, respiration control ratio, and ATP content and had further deteriorated cardiac malfunction. In conclusion, RIP140 overexpression leads to comparable cardiac function as resulted from MI, but RIP140 aggravates metabolic repression, mitochondrial malfunction, and further accelerates the transition to HF in response to MI stress.


Subject(s)
Heart Failure/complications , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Adenosine Triphosphate/metabolism , Adenoviridae/metabolism , Animals , Cell Respiration , Chronic Disease , Electrocardiography , Gene Expression Regulation , Gene Regulatory Networks , Genetic Vectors/metabolism , Heart Failure/genetics , Heart Failure/physiopathology , Hemodynamics , Male , Mitochondria, Heart/ultrastructure , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Myocardium/ultrastructure , Organelle Biogenesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Ventricular Dysfunction, Left
SELECTION OF CITATIONS
SEARCH DETAIL
...