Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Tumori ; 109(3): 276-281, 2023 Jun.
Article in English | MEDLINE | ID: mdl-35723064

ABSTRACT

OBJECTIVE: This study aimed to explore erb-b2 receptor tyrosine kinase 2 (ERBB2) gene mutations in patients with non-small cell lung cancers (NSCLC) in Northeast China, and to analyze ERBB2 mutation subtypes and clinicopathological characteristics related to the presence of ERBB2 mutations. METHODS: In this study, 1087 tissue samples, 368 whole blood samples, and 68 pleural effusion samples from 1349 NSCLC were collected. Next-generation sequencing (NGS) was used to perform genetic testing on the samples. The proportion of patients with ERBB2 mutations and related clinicopathological characteristics were analyzed. RESULTS: The mutation rate of ERBB2 in NSCLC was 5.58% (85/1523). Of the patients with ERBB2 mutations, 27.63% (21/76) were over 65 years old, 59.21% (45/76) were women, and 68.42% (52/76) were non-smokers. The majority of tumors were adenocarcinomas (92.1%, 70/76) and stage III and IV diseases accounted for 81.58% (62/76) of all cases. There were 14 subtypes of ERBB2 mutations; the most frequently seen were ERBB2 copy number alteration (41.76%, 38/91) and ERBB2 exon 20 in-frame insertion (36.26%, 33/91). Of the patients with ERBB2 mutations, 24 had concurrent epidermal growth factor receptor mutations, seven had mesenchymal epithelial transition factor amplifications, and three had anaplastic lymphoma kinase mutations. The agreement between tissue and paired blood samples in the presence of ERBB2 mutations was 64.3% (9/14). CONCLUSION: ERBB2 mutations in Northeast China NSCLC patients have a unique molecular spectrum. Our work can provide guidance for the clinical diagnosis and treatment of patients with ERBB2 mutations in Northeast China.


Subject(s)
Adenocarcinoma , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Receptor, ErbB-2 , Aged , Female , Humans , Male , Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/pathology , China/epidemiology , Lung Neoplasms/pathology , Mutation , Receptor, ErbB-2/genetics
2.
Transl Lung Cancer Res ; 12(12): 2538-2549, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38205210

ABSTRACT

Background: Anaplastic lymphoma kinase (ALK) rearrangement is one of the most important drivers in non-small cell lung cancer (NSCLC). Despite the effectiveness to canonical 3'-ALK fusions, the clinical efficacy of ALK inhibitors in patients with complex ALK fusions, such as nonreciprocal/reciprocal translocation remains uncertain. Exploring the optimal therapeutic regimens for this subset of patients is of crucial clinical significance. Case Description: We reported a female patient diagnosed with stage IVB lung adenocarcinoma (LUAD) harboring a novel ALK-RNF144A fusion, concurrent with a Huntingtin-interacting protein 1 (HIP1)-ALK fusion and a RB1 loss-of-function variant. The patient sequentially received multiple lines of treatment with ALK-tyrosine kinase inhibitor (TKI), chemotherapy, radiotherapy and ALK-TKI combined with anti-angiogenesis. Disease progression accompanied by a squamous cell carcinoma transformation was indicated after ALK-TKI combined with anti-angiogenesis and both ALK-RNF144A and HIP1-ALK fusions were retained in the tumor. The patient was subsequently treated with a third generation ALK-TKI, lorlatinib, in combination with albumin-bound paclitaxel and anlotinib, and then achieved stable disease. The patient remained on the treatment as of the last follow-up resulting in an overall survival (OS) of more than 18 months. Conclusions: We have reported an advanced NSCLC patient with a complex nonreciprocal/reciprocal ALK translocation containing a novel ALK-RNF144A fusion, concurrent with a RB1 loss-of-function mutation, who subsequently experienced pathological squamous cell carcinoma transformation. The combined treatment with ALK-TKI, chemotherapy, and anti-angiogenesis demonstrates clinical efficacy and may provide optional therapeutic strategies for this phenotype.

3.
Brain Sci ; 12(9)2022 Aug 27.
Article in English | MEDLINE | ID: mdl-36138881

ABSTRACT

Glioblastoma is the most common primary malignant brain tumor, and median survival is relatively short despite aggressive standard treatment. Natural killer (NK) cell dysfunction is strongly associated with tumor recurrence and metastasis but is unclear in glioblastoma. NK activity (NKA) represents NK cell-secreted interferon-γ (IFN-γ), which modulates immunity and inhibits cancer progression. This study aimed to analyze NKA in glioblastoma patients to obtain a clearer overview of immunity surveillance. From 2020 to 2021, a total of 20 patients and six healthy controls were recruited. Peripheral blood samples were collected preoperatively and on postoperative days (POD) 3 and 30. Then, NKA was measured using the NK VUE kit. Although NKA decreased on POD3, it recovered and further significantly enhanced on POD30, with a nearly five-fold increase compared to baseline (p = 0.004). Furthermore, the percentage of CD56brightCD16- NK cells decreased significantly on POD3 (p = 0.022) and further recovered on PO30. Subgroup analysis of extent surgical resection further revealed that the recovery of impaired NKA was attributable to gross total resection (GTR) rather than subtotal resection (STR). In conclusion, NKA is significantly impaired in glioblastoma, and GTR has demonstrated superior benefit in improving the suppressed NKA and increased CD56brightCD16- NK subset in glioblastoma patients, which may be associated with subsequent patients' prognosis. Therefore, the goal of performing GTR for glioblastoma should be achieved when possible since it appears to increase NKA cell immunity.

4.
Oncol Rep ; 48(1)2022 07.
Article in English | MEDLINE | ID: mdl-35583004

ABSTRACT

Relapse and drug resistance are the main causes of mortality in patients with small­cell lung cancer (SCLC). Intratumoral heterogeneity (ITH) is a key biological mechanism that leads to relapse and drug resistance. Phenotypic plasticity is an important factor that leads to ITH in SCLC, although its mechanisms and key regulatory factors remain to be elucidated. In the present study, cell proliferation and cell switch assay were measured using trypan blue. Alamar Blue was used to test drug sensitivity. Differential genes were screened by RNA sequencing. Reverse transcription­quantitative PCR and western blotting were performed to assess the expressions of CSF2/p­STAT3/MYC pathway related molecules, neuroendocrine (NE)/non­neuroendocrine (non­NE), transcription factors and drug­related targets. The present study found that SCLC cell line NCI­H69 exhibited adherent (H69A) and suspensive (H69S) phenotypes, which could switch back and forth. The two phenotypic cells had significant differences in cellular NE and non­NE characteristics, drug sensitivity and expression of drug­related targets. RNA sequencing showed that granulocyte­macrophage colony­stimulating factor [i.e., colony­stimulating factor 2 (CSF2)] was the main differentially expressed gene between the two phenotypes and that H69A cells highly expressed CSF2. The inhibition of CSF2 promoted the transformation from H69A to H69S, increased drug sensitivity and NE marker expression and decreased the non­NE marker expression in H69A. The STRING, Pathway Commons and Reactome databases showed a potential regulatory relationship between CSF2 and phosphorylated signal transducer and activator of transcription 3 (p­STAT3)/MYC. p­STAT3 and MYC expression was higher in H69A cells than in H69S cells and CSF2 silencing inhibited their expression. Taken together, these results indicated that CSF2 may regulate the phenotypic plasticity of SCLC through the phosphorylated STAT3/MYC pathway, thereby limiting the transformation between cell clones with different phenotypes and changing the sensitivity of specific cell clones to targeted drugs. Targeting CSF2 may be a potential therapeutic strategy to overcome drug resistance in SCLC treatment by influencing ITH.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lung Neoplasms , Small Cell Lung Carcinoma , Adaptation, Physiological , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism
5.
Oncogene ; 41(20): 2833-2845, 2022 05.
Article in English | MEDLINE | ID: mdl-35418692

ABSTRACT

Dysregulation of pericellular proteolysis is strongly implicated in cancer metastasis through alteration of cell invasion and the microenvironment. Matriptase-2 (MT-2) is a membrane-anchored serine protease which can suppress prostate cancer (PCa) cell invasion. In this study, we showed that MT-2 was down-regulated in PCa and could suppress PCa cell motility, tumor growth, and metastasis. Using microarray and biochemical analysis, we found that MT-2 shifted TGF-ß action towards its tumor suppressor function by repressing epithelial-to-mesenchymal transition (EMT) and promoting Smad2 phosphorylation and nuclear accumulation to upregulate two TGF-ß1 downstream effectors (p21 and PAI-1), culminating in hindrance of PCa cell motility and malignant growth. Mechanistically, MT-2 could dramatically up-regulate the expression of nuclear receptor NR4A3 via iron metabolism in PCa cells. MT-2-induced NR4A3 further coactivated Smad2 to activate p21 and PAI-1 expression. In addition, NR4A3 functioned as a suppressor of PCa and mediated MT-2 signaling to inhibit PCa tumorigenesis and metastasis. These results together indicate that NR4A3 sustains MT-2 signaling to suppress PCa cell invasion, tumor growth, and metastasis, and serves as a contextual factor for the TGF-ß/Smad2 signaling pathway in favor of tumor suppression via promoting p21 and PAI-1 expression.


Subject(s)
DNA-Binding Proteins , Membrane Proteins , Prostatic Neoplasms , Receptors, Steroid , Receptors, Thyroid Hormone , Serine Endopeptidases , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Epithelial-Mesenchymal Transition , Humans , Male , Membrane Proteins/metabolism , Neoplasm Invasiveness , Plasminogen Activator Inhibitor 1 , Prostate/pathology , Prostatic Neoplasms/pathology , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Serine Endopeptidases/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment
6.
Technol Cancer Res Treat ; 20: 15330338211039948, 2021.
Article in English | MEDLINE | ID: mdl-34851203

ABSTRACT

Purpose: The role of different circulating lymphocyte subsets, as well as their correlation with clinical characteristics of small cell lung cancer patients have not yet been fully understood. This study aims to evaluate the influence of the fluctuating absolute numbers of lymphocyte subpopulations in peripheral blood of patients with small cell lung cancer. Methods: The absolute counts and percentages of lymphocyte subsets in peripheral blood of 329 patients with small cell lung cancer were retrospectively analyzed. The numbers of CD3+, CD3+CD4+, and CD3+CD8+ T lymphocytes, CD3-CD19+ B lymphocytes, and CD3-CD16+CD56+ NK cells were evaluated by flow cytometry. Their relationship with the patients' clinical characteristics were statistically evaluated. Results: The CD4/CD8 values derived from the absolute number and percentage of CD3+CD4+ cells divided by CD3+CD8+ cells were identical (1.86 ± 0.99). There was no association between any of the lymphocyte subsets levels and age/sex of the 329 patients with small cell lung cancer. The patients with advanced stage had a reduction in CD3+ and CD3+CD4+ T cell counts and a decreased CD4/CD8 ratio. The levels of CD3+CD4+ T cells, CD3-CD19+ B cells, CD3-CD16+CD56+ NK cells, and CD4/CD8 ratio were associated with advanced tumor-node-metastasis stage. Patients who had undergone radiotherapy were characterized by lymphopenia with lower numbers of CD3+, CD3+CD4+, CD3+CD8+ T lymphocyte, B lymphocyte, NK cell, and CD4/CD8 ratio. The evaluation of individual CD4/CD8 ratio should be combined with other clinical parameters. Conclusions: Patients with small cell lung cancer have altered lymphocyte homeostasis. Lymphopenia was a long-lasting feature of the enrolled patients who were treated with radiotherapy. The available lymphocyte subsets levels might be used to manage the clinical treatment scheme.


Subject(s)
Lung Neoplasms/immunology , Lung Neoplasms/radiotherapy , Lymphocytes , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/radiotherapy , Aged , Antigens, CD19/metabolism , B-Lymphocytes/metabolism , CD3 Complex/metabolism , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Female , Humans , Killer Cells, Natural , Lung Neoplasms/pathology , Lymphocytes/radiation effects , Lymphopenia/etiology , Male , Middle Aged , Neoplasm Staging , Radiotherapy/adverse effects , Small Cell Lung Carcinoma/secondary , T-Lymphocytes, Cytotoxic , T-Lymphocytes, Helper-Inducer
7.
Cancer Res ; 81(12): 3270-3282, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33771897

ABSTRACT

Current clinical trials of combined EGFR-tyrosine kinase inhibitors (TKI) and immune checkpoint blockade (ICB) therapies show no additional effect. This raises questions regarding whether EGFR-TKIs attenuate ICB-enhanced CD8+ T lymphocyte function. Here we show that the EGFR-TKI afatinib suppresses CD8+ T lymphocyte proliferation, and we identify CAD, a key enzyme of de novo pyrimidine biosynthesis, to be a novel afatinib target. Afatinib reduced tumor-infiltrating lymphocyte numbers in Lewis lung carcinoma (LLC)-bearing mice. Early afatinib treatment inhibited CD8+ T lymphocyte proliferation in patients with non-small cell lung cancer, but their proliferation unexpectedly rebounded following long-term treatment. This suggests a transient immunomodulatory effect of afatinib on CD8+ T lymphocytes. Sequential treatment of afatinib with anti-PD1 immunotherapy substantially enhanced therapeutic efficacy in MC38 and LLC-bearing mice, while simultaneous combination therapy showed only marginal improvement over each single treatment. These results suggest that afatinib can suppress CD8+ T lymphocyte proliferation by targeting CAD, proposing a timing window for combined therapy that may prevent the dampening of ICB efficacy by EGFR-TKIs. SIGNIFICANCE: This study elucidates a mechanism of afatinib-mediated immunosuppression and provides new insights into treatment timing for combined targeted therapy and immunotherapy. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3270/F1.large.jpg.


Subject(s)
Afatinib/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Deoxyribonucleases/antagonists & inhibitors , Immunomodulating Agents/pharmacology , Pyrimidines/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Drug Therapy, Combination , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors
8.
Onco Targets Ther ; 13: 11499-11504, 2020.
Article in English | MEDLINE | ID: mdl-33204104

ABSTRACT

PURPOSE: The rearrangement of ROS1 (C-ros oncogene 1) is an important driver of non-small cell lung cancer (NSCLC). Currently, only approximately 24 ROS1 fusion partners have been shown to be sensitive to crizotinib. Although fusion partner determination is not required to treat patients with tyrosine kinase inhibitor, the correlation between ROS1 phenotypes and efficacies still needs more researches. Furthermore, non-reciprocal/reciprocal ROS1 translocations are rare and have not yet been reported. Thus, more novel ROS1 fusion partners and non-reciprocal/reciprocal fusions need to be provided and supplemented to guide targeted therapy and prognosis for patients. CASE PRESENTATION: Targeted next-generation sequencing panel was used to identify ROS1 rearrangements in a Chinese patient with advanced lung adenocarcinoma. We identified a non-reciprocal/reciprocal ROS1 translocation which contained a novel ROS1-FBXL17 (F-box and leucine-rich repeat protein 17) fusion co-existing with the CD74-ROS1 fusion and the patient was sensitive to crizotinib. The ROS1 rearrangement was then validated using RT-qPCR. The progression-free survival (PFS) was 15.7 months which exceeded the highest PFS level (14.2 months) in the Chinese population reported recently. Thus, this non-reciprocal/reciprocal ROS1 translocation patient had an excellent efficacy to crizotinib which was different from that in ALK. And it may be possible that the ROS1-FBXL17 fusion in this patient synergistically promotes the sensitivity of the CD74-RSO1 fusion to crizotinib. CONCLUSION: The ROS1-FBXL17 fusion may be a novel driver of NSCLC and we provide a non-reciprocal/reciprocal ROS1 translocation mode very sensitive to crizotinib. Our study adds new data to the ROS1 fusion database and provides a reference strategy for the clinical treatment of patients with double ROS1 fusions or non-reciprocal/reciprocal ROS1 translocation.

9.
Oncogene ; 39(37): 5950-5963, 2020 09.
Article in English | MEDLINE | ID: mdl-32778768

ABSTRACT

TMPRSS2 is an important membrane-anchored serine protease involved in human prostate cancer progression and metastasis. A serine protease physiologically often comes together with a cognate inhibitor for execution of proteolytically biologic function; however, TMPRSS2's cognate inhibitor is still elusive. To identify the cognate inhibitor of TMPRSS2, in this study, we applied co-immunoprecipitation and LC/MS/MS analysis and isolated hepatocyte growth factor activator inhibitors (HAIs) to be potential inhibitor candidates for TMPRSS2. Moreover, the recombinant HAI-2 proteins exhibited a better inhibitory effect on TMPRSS2 proteolytic activity than HAI-1, and recombinant HAI-2 proteins had a high affinity to form a complex with TMPRSS2. The immunofluorescence images further showed that TMPRSS2 was co-localized to HAI-2. Both KD1 and KD2 domain of HAI-2 showed comparable inhibitory effects on TMPRSS2 proteolytic activity. In addition, HAI-2 overexpression could suppress the induction effect of TMPRSS2 on pro-HGF activation, extracellular matrix degradation and prostate cancer cell invasion. We further determined that the expression levels of TMPRSS2 were inversely correlated with HAI-2 levels during prostate cancer progression. In orthotopic xenograft animal model, TMPRSS2 overexpression promoted prostate cancer metastasis, and HAI-2 overexpression efficiently blocked TMPRSS2-induced metastasis. In summary, the results together indicate that HAI-2 can function as a cognate inhibitor for TMPRSS2 in human prostate cancer cells and may serve as a potential factor to suppress TMPRSS2-mediated malignancy.


Subject(s)
Membrane Glycoproteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Serine Endopeptidases/metabolism , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Disease Models, Animal , Heterografts , Humans , Male , Membrane Glycoproteins/chemistry , Neoplasm Invasiveness , Prostatic Neoplasms/etiology , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Proteinase Inhibitory Proteins, Secretory/metabolism , Proteolysis
13.
Br J Cancer ; 120(5): 499-511, 2019 03.
Article in English | MEDLINE | ID: mdl-30765871

ABSTRACT

BACKGROUND: Dysregulation of pericellular proteolysis usually accounts for cancer cell invasion and metastasis. Isolation of a cell-surface protease system for lung cancer metastasis is an important issue for mechanistic studies and therapeutic target identification. METHODS: Immunohistochemistry of a tissue array (n = 64) and TCGA database (n = 255) were employed to assess the correlation between serine protease inhibitors (SPIs) and lung adenocarcinoma progression. The role of SPI in cell motility was examined using transwell assays. Pulldown and LC/MS/MS were performed to identify the SPI-modulated novel protease(s). A xenografted mouse model was harnessed to demonstrate the role of the SPI in lung cancer metastasis. RESULTS: Hepatocyte growth factor activator inhibitor-2 (HAI-2) was identified to be downregulated following lung cancer progression, which was related to poor survival and tumour invasion. We further isolated a serum-derived serine protease, plasmin, to be a novel target of HAI-2. Downregulation of HAI-2 promotes cell surface plasmin activity, EMT, and cell motility. HAI-2 can suppress plasmin-mediated activations of HGF and TGF-ß1, EMT and cell invasion. In addition, downregulated HAI-2 increased metastasis of lung adenocarcinoma via upregulating plasmin activity. CONCLUSION: HAI-2 functions as a novel inhibitor of plasmin to suppress lung cancer cell motility, EMT and metastasis.


Subject(s)
Adenocarcinoma of Lung/metabolism , Fibrinolysin/metabolism , Lung Neoplasms/metabolism , Membrane Glycoproteins/metabolism , A549 Cells , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/secondary , Animals , Cell Line, Tumor , Cell Movement , Disease Progression , Epithelial-Mesenchymal Transition , Fibrinolysin/antagonists & inhibitors , Hepatocyte Growth Factor/metabolism , Humans , Lung Neoplasms/pathology , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Transforming Growth Factor beta1/metabolism
14.
Sci Rep ; 7(1): 15101, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118397

ABSTRACT

Dysregulation of pericellular proteolysis is often required for tumor invasion and cancer progression. It has been shown that down-regulation of hepatocyte growth factor activator inhibitor-2 (HAI-2) results in activation of matriptase (a membrane-anchored serine protease), human prostate cancer cell motility and tumor growth. In this study, we further characterized if HAI-2 was a cognate inhibitor for matriptase and identified which Kunitz domain of HAI-2 was required for inhibiting matriptase and human prostate cancer cell motility. Our results show that HAI-2 overexpression suppressed matriptase-induced prostate cancer cell motility. We demonstrate that HAI-2 interacts with matriptase on cell surface and inhibits matriptase proteolytic activity. Moreover, cellular HAI-2 harnesses its Kunitz domain 1 (KD1) to inhibit matriptase activation and prostate cancer cell motility although recombinant KD1 and KD2 of HAI-2 both show an inhibitory activity and interaction with matriptase protease domain. The results together indicate that HAI-2 is a cognate inhibitor of matriptase, and KD1 of HAI-2 plays a major role in the inhibition of cellular matritptase activation as well as human prostate cancer invasion.


Subject(s)
Cell Movement , Membrane Glycoproteins/metabolism , Protein Domains , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Male , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice, Inbred BALB C , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteolysis , RNA Interference , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/metabolism
15.
J Cell Biochem ; 117(4): 881-93, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26365534

ABSTRACT

Ketamine, a dissociative anesthetic, is misused and abused worldwide as an illegal recreational drug. In addition to its neuropathic toxicity, ketamine abuse has numerous effects, including renal failure; however, the underlying mechanism is poorly understood. The process called epithelial phenotypic changes (EPCs) causes the loss of cell-cell adhesion and cell polarity in renal diseases, as well as the acquisition of migratory and invasive properties. Madin-Darby canine kidney cells, an in vitro cell model, were subjected to experimental manipulation to investigate whether ketamine could promote EPCs. Our data showed that ketamine dramatically decreased transepithelial electrical resistance and increased paracellular permeability and junction disruption, which were coupled to decreased levels of apical junctional proteins (ZO-1, occludin, and E-cadherin). Consistent with the downregulation of epithelial markers, the mesenchymal markers N-cadherin, fibronectin, and vimentin were markedly upregulated following ketamine stimulation. Of the E-cadherin repressor complexes tested, the mRNA levels of Snail, Slug, Twist, and ZEB1 were elevated. Moreover, ketamine significantly enhanced migration and invasion. Ketamine-mediated changes were at least partly caused by the inhibition of GSK-3ß activity through Ser-9 phosphorylation by the PI3K/Akt pathway. Inhibiting PI3K/Akt with LY294002 reactivated GSK-3ß and suppressed ketamine-enhanced permeability, EPCs, and motility. These findings were recapitulated by the inactivation of GSK-3ß using the inhibitor 3F8. Taken together, these results provide evidence that ketamine induces renal distal tubular EPCs through the downregulation of several junction proteins, the upregulation of mesenchymal markers, the activation of Akt, and the inactivation of GSK-3ß.


Subject(s)
Analgesics/pharmacology , Cell Membrane/drug effects , Epithelial-Mesenchymal Transition/drug effects , Glycogen Synthase Kinase 3/genetics , Ketamine/pharmacology , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Cell Polarity/drug effects , Dogs , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Intercellular Junctions/drug effects , Madin Darby Canine Kidney Cells , Occludin/genetics , Occludin/metabolism , Phenotype , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Vimentin/genetics , Vimentin/metabolism , Zinc Fingers/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
16.
Cell Tissue Res ; 362(1): 69-86, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25948482

ABSTRACT

The use of human mesenchymal stem cells (hMSCs) in cell therapies has increased the demand for strategies that allow efficient cell scale-up. Preliminary data on the three-dimensional (3D) spinner culture describing the potential use of microcarriers for hMSCs culture scale-up have been reported. We exploited a rich source of autologous stem cells (human hair follicle) and demonstrated the robust in vitro long-term expansion of human hair follicle-derived mesenchymal stem cells (hHF-MSCs) by using CultiSpher(®)-G microcarriers. We analyzed the feasibility of 3D culture by using hHF-MSCs/CultiSpher(®)-G microcarrier constructs for its potential applicability in regenerative medicine by comparatively analyzing the performance of hHF-MSCs adhered to the CultiSpher(®)-G microspheres in 3D spinner culture and those grown on the gelatin-coated plastic dishes (2D culture), using various assays. We showed that the hHF-MSCs seeded at various densities quickly adhered to and proliferated well on the microspheres, thus generating at least hundreds of millions of hHF-MSCs on 1 g of CultiSpher(®)-G within 12 days. This resulted in a cumulative cell expansion of greater than 26-fold. Notably, the maximum and average proliferation rates in 3D culture were significantly greater than that of the 2D culture. However, the hHF-MSCs from both the cultures retained surface marker and nestin expression, proliferation capacity and differentiation potentials toward adipocytes, osteoblasts and smooth muscle cells and showed no significant differences as evidenced by Edu incorporation, cell cycle, colony formation, apoptosis, biochemical quantification and qPCR assays.


Subject(s)
Hair Follicle/metabolism , Mesenchymal Stem Cells/metabolism , Adult , Cell Differentiation , Cell Proliferation , Female , Hair Follicle/cytology , Humans , Male , Middle Aged , Regenerative Medicine
17.
Cancer Res ; 75(14): 2949-60, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26018085

ABSTRACT

Dysregulation of androgen signaling and pericellular proteolysis is necessary for prostate cancer progression, but the links between them are still obscure. In this study, we show how the membrane-anchored serine protease TMPRSS2 stimulates a proteolytic cascade that mediates androgen-induced prostate cancer cell invasion, tumor growth, and metastasis. We found that matriptase serves as a substrate for TMPRSS2 in mediating this proinvasive action of androgens in prostate cancer. Further, we determined that higher levels of TMPRSS2 expression correlate with higher levels of matriptase activation in prostate cancer tissues. Lastly, we found that the ability of TMPRSS2 to promote prostate cancer tumor growth and metastasis was associated with increased matriptase activation and enhanced degradation of extracellular matrix nidogen-1 and laminin ß1 in tumor xenografts. In summary, our results establish that TMPRSS2 promotes the growth, invasion, and metastasis of prostate cancer cells via matriptase activation and extracellular matrix disruption, with implications to target these two proteases as a strategy to treat prostate cancer.


Subject(s)
Androgens/pharmacology , Extracellular Matrix/metabolism , Prostatic Neoplasms/pathology , Serine Endopeptidases/metabolism , Serine Endopeptidases/physiology , Animals , CHO Cells , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cricetinae , Cricetulus , Enzyme Activation/drug effects , Enzyme Activation/genetics , Extracellular Matrix/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Serine Endopeptidases/genetics , Tumor Cells, Cultured , Up-Regulation/drug effects , Up-Regulation/genetics
18.
Stem Cell Res Ther ; 6: 38, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25889402

ABSTRACT

INTRODUCTION: Successful stem cell therapy relies on large-scale generation of stem cells and their maintenance in a proliferative multipotent state. This study aimed to establish a three-dimension culture system for large-scale generation of hWJ-MSC and investigated the self-renewal activity, genomic stability and multi-lineage differentiation potential of such hWJ-MSC in enhancing skin wound healing. METHODS: hWJ-MSC were seeded on gelatin microbeads and cultured in spinning bottles (3D). Cell proliferation, karyotype analysis, surface marker expression, multipotent differentiation (adipogenic, chondrogenic, and osteogenic potentials), and expression of core transcription factors (OCT4, SOX2, NANOG, and C-MYC), as well as their efficacy in accelerating skin wound healing, were investigated and compared with those of hWJ-MSC derived from plate cultres (2D), using in vivo and in vitro experiments. RESULTS: hWJ-MSC attached to and proliferated on gelatin microbeads in 3D cultures reaching a maximum of 1.1-1.30×10(7) cells on 0.5 g of microbeads by days 8-14; in contrast, hWJ-MSC derived from 2D cultures reached a maximum of 6.5 -11.5×10(5) cells per well in a 24-well plate by days 6-10. hWJ-MSC derived by 3D culture incorporated significantly more EdU (P<0.05) and had a significantly higher proliferation index (P<0.05) than those derived from 2D culture. Immunofluorescence staining, real-time PCR, flow cytometry analysis, and multipotency assays showed that hWJ-MSC derived from 3D culture retained MSC surface markers and multipotency potential similar to 2D culture-derived cells. 3D culture-derived hWJ-MSC also retained the expression of core transcription factors at levels comparable to their 2D culture counterparts. Direct injection of hWJ-MSC derived from 3D or 2D cultures into animals exhibited similar efficacy in enhancing skin wound healing. CONCLUSIONS: Thus, hWJ-MSC can be expanded markedly in gelatin microbeads, while retaining MSC surface marker expression, multipotent differential potential, and expression of core transcription factors. These cells also efficiently enhanced skin wound healing in vivo, in a manner comparable to that of hWJ-MSC obtained from 2D culture.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cells/cytology , Skin/injuries , Wharton Jelly/cytology , Wound Healing/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation , Gelatin/metabolism , Homeodomain Proteins/biosynthesis , Humans , Mice , Microspheres , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Tissue Culture Techniques
19.
Fundam Clin Pharmacol ; 29(2): 191-203, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25619112

ABSTRACT

Cyclosporine A (CsA) enhances hair growth through caspase-dependent pathways by retarding anagen-to-catagen phase transition in the hair follicle growth cycle. Whether apoptosis-inducing factor (AIF), a protein that induces caspase-independent apoptosis, can regulate the hair follicle cycle in response to CsA is currently unclear. Here, we show that the pro-hair growth properties of CsA are in part due to blockage of AIF nuclear translocation. We first isolate hair follicles from murine dorsal skin. We then used Western blot, immunohistochemistry and immunofluorescence to evaluate the expression and localization of AIF in hair follicles. We also determined whether modulation of AIF was responsible for the effects of CsA at the anagen-to-catagen transition. AIF was expressed in hair follicles during the anagen, catagen and telogen phases. There was significant nuclear translocation of AIF as hair follicles transitioned from anagen to late catagen phase; this was inhibited by CsA, likely due to reduced cyclophilin A expression and attenuated AIF release from mitochondria. However, we note that AIF translocation was not completely eliminated, which likely explains why the transition to catagen phase was severely retarded by CsA, rather than being completely inhibited. We speculate that blockade of the AIF signalling pathway is a critical event required for CsA-dependent promotion of hair growth in mice. The study of AIF-related signalling pathways may provide insight into hair diseases and suggest potential novel therapeutic strategies.


Subject(s)
Apoptosis Inducing Factor/metabolism , Cell Nucleus/metabolism , Cyclosporine/pharmacology , Hair Follicle/metabolism , Animals , Apoptosis Inducing Factor/antagonists & inhibitors , Cell Nucleus/drug effects , Hair Follicle/drug effects , Mice , Mice, Inbred C57BL
20.
Cell Transplant ; 24(5): 891-907, 2015.
Article in English | MEDLINE | ID: mdl-24835482

ABSTRACT

Genetically engineered stem cells that overexpress genes encoding therapeutic products can be exploited to correct metabolic disorders by repairing and regenerating diseased organs or restoring their function. Hair follicles are readily accessible and serve as a rich source of autologous stem cells for cell-based gene therapy. Here we isolated mesenchymal stem cells from human hair follicles (HF-MSCs) and engineered them to overexpress the human insulin gene and release human insulin in a time- and dose-dependent manner in response to rapamycin. The engineered HF-MSCs retained their characteristic cell surface markers and retained their potential to differentiate into adipocytes and osteoblasts. When mice with streptozotocin-induced type 1 diabetes were engrafted with these engineered HF-MSCs, these cells expressed and released a dose of human insulin, dramatically reversed hyperglycemia, and significantly reduced death rate. Moreover, the engineered HF-MSCs did not form detectable tumors throughout the 120-day animal tests in our experiment. Our results show that HF-MSCs can be used to safely and efficiently express therapeutic transgenes and therefore show promise for cell-based gene therapy of human disease.


Subject(s)
Cell Engineering , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Hair Follicle/metabolism , Hyperglycemia/therapy , Insulin/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Adult , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Female , Heterografts , Humans , Hyperglycemia/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...