Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
World J Clin Cases ; 10(21): 7365-7375, 2022 Jul 26.
Article in English | MEDLINE | ID: mdl-36157997

ABSTRACT

BACKGROUND: It is estimated that about 30% of esophageal cancer (EC) patients are over 70 years old. Therefore, there is less evidence on the diagnosis and management of elderly EC patients. It is important to explore how elderly EC patients benefit from radical radiochemotherapy regimens, including the target area of radiotherapy (RT), radiation dose and fraction, and choice of chemotherapy drugs. AIM: To compare the efficacy of involved-field intensity-modulated RT (IF-IMRT) combined with S-1 vs RT alone in the treatment of elderly EC patients in terms of safety, short-term response, and survival. METHODS: Thirty-four EC patients aged > 70 years were prospectively enrolled between December 2017 and December 2019. Based on the random number table, they were divided into an IF-IMRT + S-1 group and an IF-IMRT alone group, with 17 patients in each group. All patients were treated with IF-IMRT at a dose of 50.4-56 Gy in 28-30 fractions (1.8-2 Gy/fraction, 5 fractions/wk). Oral S-1 was administered concomitantly in the IF-IMRT + S-1 group for 14 consecutive days, and a second cycle was started 7 d after drug withdrawal. After RT, 4 cycles of S-1 treatment were offered as the consolidation chemotherapy. The safety, short-term response, and survival were observed after the treatment. RESULTS: As of April 2022, these 34 patients had been followed up for 15.2-32.5 mo, with a median follow-up period of 24.5 mo. Complete efficacy indicators were obtained from all the patients. The objective response rate was 88.2% vs 76.5%, respectively, in the IF-IMRT + S-1 group and the RT alone group, where as the disease control rate was 100% vs 82.4%, respectively. The incidence of adverse events including grade 1-2 fatigue, granulocytopenia, thrombocytopenia, anemia, radiation esophagitis, radiation-induced skin injury, and radiation-induced lung injury was not significantly different between these two groups, so was the incidence of the grade 3 radiation esophagitis (0% vs 5.7%). The rate of progressive disease (PD) was 52.9% (n = 9) in the IF-IMRT + S-1 group and 64.7% (n = 11) in the RT alone group. The median progression-free survival (PFS) was 23.4 mo vs 16.3 mo, and the 2-year PFS rate was 42% vs 41.2%. The median overall survival (OS) was 27.0 mo vs 23.0 mo, and the 2-year OS rate was 58.8% vs 47.1%. Multivariate analysis showed that age was a significant prognostic factor (P = 0.0019); patients aged < 75 years had a significant survival advantage over patients aged ≥ 75 years. The locations of EC also affected the prognosis. In the IF-IMRT + S-1 group, the number of chemotherapy cycles was a significant prognostic factor (P = 0.0125), and the risk of PD was significantly lower in EC patients who had received 6 cycles of chemotherapy than those who had received 2-5 cycles of chemotherapy. CONCLUSION: Compared with IF-IMRT alone, IF-IMRT + S-1 shows the benefits of preventing PD and prolonging survival without increasing adverse reactions. Therefore, this concurrent radiochemotherapy deserves clinical application.

2.
Biomed Environ Sci ; 32(9): 673-686, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31635683

ABSTRACT

OBJECTIVE: Cancer is a serious threat to human health. Despite extensive research on cancer treatment, there is a growing demand for new therapies. CD147 is widely involved in tumor development, but it is unclear whether cancer cell malignancy is affected by CD147 expression level. The first compound (AC-73) targeting CD147 could only act on advanced tumors and inhibit metastasis. Therefore, new compounds with better anticancer activity should be explored. METHODS: Wst-1 assays were used to confirm the effect of novel compounds on proliferation. Apoptosis tests were used to evaluate their proapoptotic capacity. A nude mouse model was used to demonstrate in vivo anticancer activity and safety of the compounds. Western blots were used to suggest a molecule mechanism. RESULTS: There is a positive correlation between CD147 expression and tumor cell proliferation. A new compound, HA-08, was synthesized and proved to be more active than AC-73. HA-08 could inhibit cancer cell viability and promote cancer cell apoptosis both in vitro and in vivo. HA-08 induces cancer apoptosis, mainly by disrupting the CD147-CD44 interaction and then down-regulating the JAK/STAT3/Bcl-2 signaling pathway. CONCLUSION: Our results have clarified the tumor specificity of CD147 and its drug target characteristics. The biological profile of HA-08 suggests that this compound could be developed as a potential anticancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Basigin/antagonists & inhibitors , Cell Survival/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Mice , Mice, Nude
3.
Int J Nanomedicine ; 14: 623-638, 2019.
Article in English | MEDLINE | ID: mdl-30697046

ABSTRACT

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignancies, with high rates of mortality and morbidity worldwide. Owing to the special anatomical location of this tumor, an effective, minimally invasive treatment with low systemic toxicity is highly desirable. Hydrogels have shown great potential for tumor-targeting therapy, with excellent performance. However, there have been few reports on co-loading photosensitizers and chemotherapeutic drugs into hydrogels. In this study, we synthesized a nano doxorubicin-indocyanine green matrix metalloproteinase (MMP)-responsive hydrogel (denoted as NDIMH), combining chemotherapy and phototherapy, to achieve superior antitumor efficacy. METHODS: First, NDIMH was synthesized and characterized by scanning electron microscopy and drug-release assays. Second, the photosensitivity properties and antitumor efficiency of this drug delivery system were studied in vivo and in vitro. Last, the imaging and biodistribution of NDIMH were monitored using the Maestro EX in vivo imaging system. RESULTS: The nanodrugs loaded into the smart hydrogel exhibited uniform size distribution, excellent size stability, and a sustained release in the presence of MMP-2. NDIMH showed ideal photosensitivity characteristics under light. NDIMH with 808 nm near-infrared (NIR) irradiation effectively inhibited the viability, invasion, and metastasis of SCC-15 in vitro. After intratumoral injection of NDIMH with 808 nm NIR illumination, the hydrogels exhibited favorable synergistic antitumor efficacy and acceptable biosafety. Additionally, fluorescence imaging showed that NDIMH could significantly improve the retention of nanodrugs at the tumor site. CONCLUSION: The intratumoral injection of NDIMH with 808 nm NIR irradiation could be a promising chemophototherapy alternative for HNSCC.


Subject(s)
Doxorubicin/therapeutic use , Hydrogels/chemical synthesis , Indocyanine Green/therapeutic use , Matrix Metalloproteinases/metabolism , Nanoparticles/chemistry , Photochemotherapy , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Doxorubicin/pharmacology , Fluorescence , Humans , Hydrogels/chemistry , Indocyanine Green/pharmacology , Light , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , Neoplasm Invasiveness , Neoplasm Metastasis , Rats , Tissue Distribution
4.
Sci Rep ; 6: 35210, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27869218

ABSTRACT

Cluster of differentiation 147 (CD147), also known as extracellular matrix metalloproteinase inducer, is a transmembrane glycoprotein that mediates oncogenic processes partly through N-glycosylation modifications. N-glycosylation has been demonstrated to be instrumental for the regulation of CD147 function during malignant transformation. However, the role that site-specific glycosylation of CD147 plays in its defective function in hepatocellular carcinomacells needs to be determined. Here, we demonstrate that the modification of N-glycosylation at Asn152 on CD147 strongly promotes hepatocellular carcinoma (HCC) invasion and migration. After the removal of N-glycans at Asn152, CD147 was more susceptible to degradation by ER-localized ubiquitin ligase-mediated endoplasmic reticulum-associated degradation (ERAD). Furthermore, N-linked glycans at Asn152 were required for CD147 to acquire and maintain proper folding in the ER. Moreover, N-linked glycans at Asn152 functioned as a recognition motif that was directly mediated by the CNX quality control system. Two phases in the retention-based ER chaperones system drove ER-localized CD147 trafficking to degradation. Deletion of N-linked glycosylation at Asn152 on CD147 significantly suppressed in situ tumour metastasis. These data could potentially shed light on the molecular regulation of CD147 through glycosylation and provide a valuable means of developing drugs that target N-glycans at Asn152 on CD147.


Subject(s)
Asparagine/chemistry , Basigin/chemistry , Carcinoma, Hepatocellular/genetics , Endoplasmic Reticulum-Associated Degradation , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Animals , Asparagine/immunology , Basigin/genetics , Basigin/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Glycosylation , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Polysaccharides/chemistry , Polysaccharides/immunology , Protein Folding , Proteolysis
5.
Oncotarget ; 7(38): 62177-62193, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27556697

ABSTRACT

Pancreatic cancer, one of the most lethal cancers, has very poor 5-year survival partly due to gemcitabine resistance. Recently, it was reported that chemotherapeutic agents may act as stressors to induce adaptive responses and to promote chemoresistance in cancer cells. During long-term drug treatment, the minority of cancer cells survive and acquire an epithelial-mesenchymal transition phenotype with increased chemo-resistance and metastasis. However, the short-term response of most cancer cells remains unclear. This study aimed to investigate the short-term response of pancreatic cancer cells to gemcitabine stress and to explore the corresponding mechanism. Our results showed that gemcitabine treatment for 24 hours enhanced pancreatic cancer cell invasion. In gemcitabine-treated cells, HAb18G/CD147 was up-regulated; and HAb18G/CD147 down-regulation or inhibition attenuated gemcitabine-enhanced invasion. Mechanistically, HAb18G/CD147 promoted gemcitabine-enhanced invasion by activating the EGFR (epidermal growth factor receptor)-STAT3 (signal transducer and activator of transcription 3) signaling pathway. Inhibition of EGFR-STAT3 signaling counteracted gemcitabine-enhanced invasion, and which relied on HAb18G/CD147 levels. In pancreatic cancer tissues, EGFR was highly expressed and positively correlated with HAb18G/CD147. These data indicate that pancreatic cancer cells enhance cell invasion via activating HAb18G/CD147-EGFR-pSTAT3 signaling. Our findings suggest that inhibiting HAb18G/CD147 is a potential strategy for overcoming drug stress-associated resistance in pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Pancreatic Neoplasms/drug therapy , Signal Transduction/drug effects , Adenocarcinoma/pathology , Animals , Antimetabolites, Antineoplastic/therapeutic use , Basigin/genetics , Basigin/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Down-Regulation , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/metabolism , Female , HEK293 Cells , Humans , Immunohistochemistry , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasm Invasiveness/pathology , Pancreas/pathology , Pancreatic Neoplasms/pathology , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Tissue Array Analysis , Up-Regulation , Xenograft Model Antitumor Assays , Gemcitabine
6.
Oncotarget ; 7(8): 9429-47, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26882566

ABSTRACT

CD147, a type I transmembrane glycoprotein, is highly expressed in various cancer types and plays important roles in tumor progression, especially by promoting the motility and invasion of hepatocellular carcinoma (HCC) cells. These crucial roles make CD147 an attractive target for therapeutic intervention in HCC, but no small-molecule inhibitors of CD147 have been developed to date. To identify a candidate inhibitor, we used a pharmacophore model derived from the structure of CD147 to virtually screen over 300,000 compounds. The 100 highest-ranked compounds were subjected to biological assays, and the most potent one, dubbed AC-73 (ID number: AN-465/42834501), was studied further. We confirmed that AC-73 targeted CD147 and further demonstrated it can specifically disrupt CD147 dimerization. Moreover, molecular docking and mutagenesis experiments showed that the possible binding sites of AC-73 on CD147 included Glu64 and Glu73 in the N-terminal IgC2 domain, which two residues are located in the dimer interface of CD147. Functional assays revealed that AC-73 inhibited the motility and invasion of typical HCC cells, but not HCC cells that lacked the CD147 gene, demonstrating on-target action. Further, AC-73 reduced HCC metastasis by suppressing matrix metalloproteinase (MMP)-2 via down-regulation of the CD147/ERK1/2/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Finally, AC-73 attenuated progression in an orthotopic nude mouse model of liver metastasis, suggesting that AC-73 or its derivatives have potential for use in HCC intervention. We conclude that the novel small-molecule inhibitor AC-73 inhibits HCC mobility and invasion, probably by disrupting CD147 dimerization and thereby mainly suppressing the CD147/ERK1/2/STAT3/MMP-2 pathways, which are crucial for cancer progression.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Basigin/metabolism , Carcinoma, Hepatocellular/drug therapy , Cell Movement/drug effects , Drug Discovery/methods , Liver Neoplasms/drug therapy , Animals , Antineoplastic Agents/adverse effects , Basigin/drug effects , Binding Sites/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , Molecular Docking Simulation , Neoplasm Invasiveness/pathology , STAT3 Transcription Factor/metabolism
7.
Oncotarget ; 7(5): 5613-29, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26716413

ABSTRACT

The acquisition of inappropriate migratory feature is crucial for tumor metastasis. It has been suggested that CD147 and Annexin A2 are involved in regulating tumor cell movement, while the regulatory mechanisms are far from clear. In this study, we demonstrated that CD147 physically interacted with the N-terminal domain of Annexin A2 and decreased Annexin A2 phosphorylation on tyrosine 23. In vitro kinase assay showed that the I domain of CD147 was indispensable for CD147-mediated downregulation of Annexin A2 phosphorylation by Src. Furthermore, we determined that p-Annexin A2 promoted the expression of dedicator of cytokinesis 3 (DOCK3) and DOCK3 blocked ß-catenin nuclear translocation, resulting in inhibition of ß-catenin signaling. In addition, DOCK3 inhibited lamellipodium dynamics and tumor cell movement. Also, we found that ß-catenin signaling increased WAVE2 expression. Therefore, DOCK3 was characterized as a negative regulator of WAVE2 expression via inhibiting ß-catenin signaling. Our study provides the first evidence that CD147 promotes tumor cell movement and metastasis via direct interaction with Annexin A2 and DOCK3-ß-catenin-WAVE2 signaling axis.


Subject(s)
Annexin A2/metabolism , Basigin/metabolism , Carcinoma, Hepatocellular/secondary , Cell Movement , Guanine Nucleotide Exchange Factors/metabolism , Nerve Tissue Proteins/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , beta Catenin/metabolism , Animals , Annexin A2/antagonists & inhibitors , Annexin A2/genetics , Apoptosis , Basigin/chemistry , Basigin/genetics , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Surface Plasmon Resonance , Tumor Cells, Cultured , Wiskott-Aldrich Syndrome Protein Family/antagonists & inhibitors , Wiskott-Aldrich Syndrome Protein Family/genetics , Xenograft Model Antitumor Assays , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
8.
Am J Cancer Res ; 5(10): 3174-85, 2015.
Article in English | MEDLINE | ID: mdl-26693068

ABSTRACT

Necroptosis, a novel form of programmed cell death, was recently shown to be strongly associated with intestinal inflammation in mice and in pediatric patients with inflammatory bowel disease (IBD). Persistent inflammation of the colon is an important risk factor for colorectal cancer. Necrostatin-1 (Nec-1), known as a specific inhibitor of necroptosis, through preventing the receptor-interacting protein (RIP) 1 and RIP3 interaction. In the present study, the anti-inflammatory and antitumorigenic efficacy of necrostatin-1 was studied in mouse models of colitis and colitis-associated cancer (CAC). We found that in acute dextran sulfate sodium (DSS)-induced colitis, treatment with necrostatin-1 significantly suppressed colitis symptoms in mice, including weight loss, colon shortening, colonic mucosa damage and severity, and excessive production of interleukin-6. Necrostatin-1 administration inhibited the upregulation of RIP1 and RIP3 and enhanced the expression of caspase-8 in DSS-induced colitis. In addition, the anti-inflammatory effect of necrostatin-1 was confirmed by in vitro analyses. Necrostatin-1 treatment reduced the production of proinflammatory cytokine and extracellular HMGB1 release in HT-29 cells in active necroptosis. Furthermore, In a mouse model of colitis-associated tumorigenesis, necrostatin-1 administration significantly suppressed tumor growth and development through inhibiting JNK/c-Jun signaling. Taken together, these findings suggest that necrostatin-1 might be a promising therapeutic option for the treatment of colitis-associated colorectal cancer in patients with IBD.

9.
Oncotarget ; 6(1): 243-57, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25428919

ABSTRACT

Metastasis is considered a dynamic process in tumor development that is related to abnormal migration and invasion. Tumor cells can move as individual cells in two interconvertible modes: mesenchymal-type and amoeboid. Previously, we reported that the interaction between CD147 and Annexin II can inhibit the amoeboid movement in hepatocellular carcinoma (HCC) cells. However, the mechanism of CD147 involved in mesenchymal movement is still unclear. Notably, our results show overexpression of CD147 led to mesenchymal-type movement in HCC cells. Evidence indicated that the mesenchymal-type cell movement induced by CD147 was Src dependent, as observed by confocal microscopy and Rac1 activity assay. The phosphorylation of Src (pY416-Src) can be up-regulated by CD147, and this regulation is mediated by focal adhesion kinase (FAK). Next, we identified DOCK8 as a GEF for Rac1, a key molecule driving mesenchymal-type movement. We also found that Src promotes STAT3 phosphorylation and STAT3 facilitates DOCK8 transcription, thus enhancing DOCK8 expression and Rac1 activation. This study provides a novel mechanism of CD147 regulating mesenchymal-type movement in HCC cells.


Subject(s)
Basigin/metabolism , Carcinoma, Hepatocellular/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Liver Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mesenchymal Stem Cells/cytology , Microscopy, Confocal , Neoplasm Metastasis , Phosphorylation , RNA Interference , Signal Transduction , Wound Healing , rho-Associated Kinases/metabolism
10.
Int J Clin Exp Med ; 8(12): 22204-16, 2015.
Article in English | MEDLINE | ID: mdl-26885196

ABSTRACT

The identification of reliable prognostic markers that distinguish patients' status and predict therapeutic response can improve the clinical outcomes of pancreatic cancer patients. The epithelial cell adhesion molecule (EpCAM) is known to be highly expressed in cancers and serves as a prognosis factor. Generally, membranous EpCAM expression in cancer cells and its clinical significance are evaluated. However, there is also an evidence of cytoplasmic EpCAM distribution in cancer cells. Hence, we investigated which kind of the immunostaining pattern in pancreatic cancer patients was, and whether membranous or cytoplasmic immunostaining had clinical significance. We determined the cytoplasmic or membranous EpCAM expression by a well-established immunohistochemical staining protocol in 157 pairs of carcinoma and paired adjacent non-tumor pancreatic tissue samples using the EpCAM-specific antibody. Furthermore, we evaluated the relationship between tumoral EpCAM expression of resected specimens and patient's overall survival as well as other biological variables like clinical prognosis by Kaplan-Meier method and χ(2) test. We found that pancreatic cancer patients had expressed higher level of cytoplasmic EpCAM but lower level of membranous EpCAM, and their expressions were significantly correlated. Cytoplasmic EpCAM acted as a favorable prognosis factor on survival time in patients with HBV negative infection. Pancreatic cancer patients with cytoplasmic EpCAM over-expression and negative Hepatitis B virus infection might benefit further from post-surgery chemotherapy. These data suggested a potential role of cytoplasmic EpCAM in predicting patient's prognosis and determining therapeutic strategy.

11.
Biochim Biophys Acta ; 1842(9): 1770-82, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24932516

ABSTRACT

Lung interstitial fibrosis is a chronic lung disease, and few effective therapies are available to halt or reverse the progression of the disease. In murine and human lung fibrosis, the expression of CD147 is increased. However, the role of CD147 in lung fibrosis has not been identified, and it remains to be determined whether lung fibrosis would be improved by decreasing the expression of CD147. A murine bleomycin-induced lung interstitial fibrosis model was used in the experiments, and HAb18 mAbs and CsA were administered during the induction of lung fibrosis. In our study, we found that the HAb18 mAbs markedly reduced the collagen score and down-regulated M1 macrophages and Th17 cells. In vitro, flow cytometry analysis showed that M1 macrophages induced higher Th17 differentiation than M2 macrophages. After treatment with HAb18 mAbs or after reducing the expression of CD147 by lentivirus interference in M1 macrophages, the level of Th17 cells were significantly inhibited. In conclusion, HAb18 mAbs or CsA treatment ameliorates lung interstitial fibrosis. CD147 promoted M1 macrophage and induced the differentiation of Th17 cells in lung interstitial fibrosis, perhaps by regulating some cytokines such as IL-6, IL-1ß, IL-12 and IL-23. These results indicated that CD147 may play an important role in the development of lung interstitial fibrosis.


Subject(s)
Basigin/metabolism , Cell Differentiation , Macrophages/metabolism , Pulmonary Fibrosis/pathology , Th17 Cells/immunology , Animals , Antibiotics, Antineoplastic/toxicity , Basigin/chemistry , Basigin/genetics , Bleomycin/toxicity , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoenzyme Techniques , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Th17 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...