Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Adv Mater ; 36(15): e2310306, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38194699

ABSTRACT

The enzymatic activities of Furin, Transmembrane serine proteinase 2 (TMPRSS2), Cathepsin L (CTSL), and Angiotensin-converting enzyme 2 (ACE2) receptor binding are necessary for the entry of coronaviruses into host cells. Precise inhibition of these key proteases in ACE2+ lung cells during a viral infection cycle shall prevent viral Spike (S) protein activation and its fusion with a host cell membrane, consequently averting virus entry to the cells. In this study, dual-drug-combined (TMPRSS2 inhibitor Camostat and CTSL inhibitor E-64d) nanocarriers (NCs) are constructed conjugated with an anti-human ACE2 (hACE2) antibody and employ Red Blood Cell (RBC)-hitchhiking, termed "Nanoengineered RBCs," for targeting lung cells. The significant therapeutic efficacy of the dual-drug-loaded nanoengineered RBCs in pseudovirus-infected K18-hACE2 transgenic mice is reported. Notably, the modular nanoengineered RBCs (anti-receptor antibody+NCs+RBCs) precisely target key proteases of host cells in the lungs to block the entry of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), regardless of virus variations. These findings are anticipated to benefit the development of a series of novel and safe host-cell-protecting antiviral therapies.


Subject(s)
COVID-19 , Cathepsin L , SARS-CoV-2 , Serine Proteinase Inhibitors , Animals , Mice , Angiotensin-Converting Enzyme 2/metabolism , Cathepsin L/antagonists & inhibitors , Cathepsin L/metabolism , COVID-19/prevention & control , COVID-19/virology , Erythrocytes , Lung/metabolism , Peptide Hydrolases/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use
2.
Adv Sci (Weinh) ; 9(22): e2201166, 2022 08.
Article in English | MEDLINE | ID: mdl-35652264

ABSTRACT

Cancer stem cells (CSCs) are reported to play essential roles in chemoresistance and metastasis. Pathways regulating CSC self-renewal and proliferation, such as Hedgehog, Notch, Wnt/ß-catenin, TGF-ß, and Myc, may be potential therapeutic targets. Here, a functional screening from the focused library with 365 compounds is performed by a step-by-step strategy. Among these candidate molecules, phenyl-2-pyrimidinyl ketone 4-allyl-3-amino selenourea (CU27) is chosen for further identification because it proves to be the most effective compound over others on CSC inhibition. Through ingenuity pathway analysis, it is shown CU27 may inhibit CSC through a well-known stemness-related transcription factor c-Myc. Gene set enrichment analysis, dual-luciferase reporter assays, expression levels of typical c-Myc targets, molecular docking, surface plasmon resonance, immunoprecipitation, and chromatin immunoprecipitation are conducted. These results together suggest CU27 binds c-Myc bHLH/LZ domains, inhibits c-Myc-Max complex formation, and prevents its occupancy on target gene promoters. In mouse models, CU27 significantly sensitizes sorafenib-resistant tumor to sorafenib, reduces the primary tumor size, and inhibits CSC generation, showing a dramatic anti-metastasis potential. Taken together, CU27 exerts inhibitory effects on CSC and CSC-associated traits in hepatocellular carcinoma (HCC) via c-Myc transcription activity inhibition. CU27 may be a promising therapeutic to treat sorafenib-resistant HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Selenium Compounds , Selenium , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Early Detection of Cancer , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Mice , Molecular Docking Simulation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Selenium/metabolism , Selenium/pharmacology , Selenium Compounds/metabolism , Selenium Compounds/pharmacology , Sorafenib/metabolism , Sorafenib/pharmacology
3.
FEBS Lett ; 595(1): 68-84, 2021 01.
Article in English | MEDLINE | ID: mdl-33040326

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common liver cancer with high mortality. Here, we found that hnRNPU is overexpressed in HCC tissues and is correlated with the poor prognosis of HCC patients. Besides, hnRNPU is of high significance in regulating the proliferation, apoptosis, self-renewal, and tumorigenic potential of HCC cells. Mechanismly, c-Myc regulates hnRNPU expression at the transcriptional level, and meanwhile, hnRNPU stabilizes the mRNA of c-MYC. We found that the hnRNPU and c-Myc regulatory loop exerts a synergistic effect on the proliferation and self-renewal of HCC, and promotes the HCC progression. Taken together, hnRNPU functions as a novel transcriptional target of c-Myc and promotes HCC progression, which may become a promising target for the treatment of c-Myc-driven HCC.


Subject(s)
Apoptosis/physiology , Carcinoma, Hepatocellular/pathology , Heterogeneous-Nuclear Ribonucleoprotein U/physiology , Liver Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic , Animals , Cell Line, Tumor , Humans , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
4.
FEBS Open Bio ; 10(9): 1737-1747, 2020 09.
Article in English | MEDLINE | ID: mdl-32662250

ABSTRACT

Liver cancer stem cells (L-CSCs) are considered to be an important therapeutic target for hepatocellular carcinoma (HCC). This study provides a new in vitro long-term culture model for a specific subpopulation of L-CSCs enriched by cell surface markers. We combined CD13, CD133 and EpCAM to selectively enrich L-CSCs, which we then cultured in modified chemically defined medium. The enriched L-CSCs exhibited enhanced proliferation, self-renewal and long-term clonal maintenance ability as compared with non-CSCs. Compared with wild-type hepatocellular carcinoma, the expression of stemness surface markers, oncogenes, drug resistance and tumorigenicity in enriched L-CSCs was significantly increased. In summary, the subpopulation of L-CSCs still maintains cancer stem cell-related phenotypes after 14 days of culture.


Subject(s)
AC133 Antigen/metabolism , Biomarkers, Tumor/metabolism , CD13 Antigens/metabolism , Carcinoma, Hepatocellular/pathology , Epithelial Cell Adhesion Molecule/metabolism , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured
5.
Cell Death Dis ; 10(6): 453, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186405

ABSTRACT

Hepatocellular carcinoma (HCC) has a high mortality rate due to the lack of effective treatments and drugs. Arsenic trioxide (ATO), which has been proved to successfully treat acute promyelocytic leukemia (APL), was recently reported to show therapeutic potential in solid tumors including HCC. However, its anticancer mechanisms in HCC still need further investigation. In this study, we demonstrated that ATO inhibits tumorigenesis and distant metastasis in mouse models, corresponding with a prolonged mice survival time. Also, ATO was found to significantly decrease the cancer stem cell (CSC)-associated traits. Minichromosome maintenance protein (MCM) 7 was further identified to be a potential target suppressed dramatically by ATO, of which protein expression is increased in patients and significantly correlated with tumor size, cellular differentiation, portal venous emboli, and poor patient survival. Moreover, MCM7 knockdown recapitulates the effects of ATO on CSCs and metastasis, while ectopic expression of MCM7 abolishes them. Mechanistically, our results suggested that ATO suppresses MCM7 transcription by targeting serum response factor (SRF)/MCM7 complex, which functions as an important transcriptional regulator modulating MCM7 expression. Taken together, our findings highlight the importance of ATO in the treatment of solid tumors. The identification of SRF/MCM7 complex as a target of ATO provides new insights into ATO's mechanism, which may benefit the appropriate use of this agent in the treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenic Trioxide/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Minichromosome Maintenance Complex Component 7/metabolism , Neoplastic Stem Cells/metabolism , Serum Response Factor/metabolism , Animals , Antineoplastic Agents/therapeutic use , Arsenic Trioxide/therapeutic use , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Disease Progression , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Ontology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/mortality , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Minichromosome Maintenance Complex Component 7/genetics , Neoplastic Stem Cells/drug effects , Prognosis , Serum Response Factor/antagonists & inhibitors , Serum Response Factor/genetics , Transplantation, Heterologous
6.
Biochem Biophys Res Commun ; 500(2): 310-317, 2018 06 02.
Article in English | MEDLINE | ID: mdl-29654765

ABSTRACT

BACKGROUND: Mesenchymal stem cell (MSC)-derived exosomes have been recognized as new candidates for the treatment of ischemic disease or injury and may be an alternative treatment for cell therapy. This aim of the study was to evaluate whether exosomes derived from adipose mesenchymal stem cell (ADSC) can protect the skin flap during ischemia-reperfusion (I/R) injury and induce neovascularization. METHODS: To investigate the effects of exosomes in the I/R injury of flap transplantation in vivo, flaps were subjected to 6 h of ischemia by ligating the left superficial inferior epigastric vessels (SIEA) followed by blood perfusion. Exosomes derived from normal ADSC (ADSC-exos) and exosomes derived from ADSC preconditioned with H2O2 (H2O2-ADSC-exos) were injected into the flaps. Then, the blood perfusion unit (BPU) of the flaps was measured by Laser Doppler Perfusion Imaging (LDPI) and microvessel density was determined by the endothelial with cell marker CD31 with Immunohistochemistry (IHC) staining. Inflammatory cell infiltration of the skin flap and apoptosis were detected by hematoxylin & eosin staining (H&E) and the TdT-mediated biotinylated dUTP nick end-labeling (TUNEL) technique. RESULTS: In vivo, exosomes significantly increased flap survival and capillary density compared to I/R on postoperative day 5, and decreased the inflammatory reaction and apoptosis in the skin flap (P < 0.05). Furthermore, H2O2-ADSC-exos had better outcomes compared to normal exosomes (P < 0.05). ADSC-exos could significantly increase human umbilical vein endothelial cell (HUVEC) proliferation (P < 0.05), but no statistic difference was found in exosomes derived from different microenvironments (P > 0.05). HUVEC co-cultured with H2O2-ADSC-exos increased the migration ratio and generated more cord-like structures compared to ADSC-exos and the control group (P < 0.05). CONCLUSION: ADSC-exos can enhance skin flap survival, promote neovascularization and alleviate the inflammation reaction and apoptosis in the skin flap after I/R injury. The use of a specific microenvironment for in vitro stem cell culture, such as one containing a low concentration of H2O2, will facilitate the development of customized exosomes for cell-free therapeutic applications in skin flap transplantation.


Subject(s)
Adipose Tissue/cytology , Exosomes/metabolism , Hydrogen Peroxide/pharmacology , Mesenchymal Stem Cells/cytology , Reperfusion Injury/pathology , Surgical Flaps/blood supply , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Endocytosis/drug effects , Exosomes/drug effects , Exosomes/transplantation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic/drug effects , Perfusion
7.
Biochem Biophys Res Commun ; 497(1): 305-312, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29428734

ABSTRACT

BACKGROUND: Adipose-derived stromal cells (ADSCs)-derived exosomes (ADSC-Exos) account for the proangiogenic potential of stem cell. This study aimed to investigate the effect of ADSC-derived exosomes (ADSC-Exos) on the survival in fat grafting. METHODS: A nude mouse model of subcutaneous fat grafting was adopted. Hypoxic preconditioned ADSC-Exos and ADSC-Exos were injected around the grafted tissue. The fat graft sample was weighed and examined by hematoxylin and eosin (H&E) staining and immunohistochemistry. Laser Doppler flowmetry and CD31 immunofluorescence staining were used to analyze neovascularization. RESULTS: ADSC-Exo and hypoxic ADSC-Exo groups had a significantly higher weight of fat graft and more perilipin-positive adipocytes than the control groups from 2 to 8 weeks after grafting, and the hypoxic ADSC-Exo group had better outcomes (all P < 0.05). H&E staining showed that ADSC-Exos attenuated infiltration of inflammatory cells around the fat grafts. Laser Doppler flowmetry showed that the two ADSC-Exo groups had better blood perfusion in the graft tissue than the control groups (all P < 0.05). Immunofluorescence demonstrated that the hypoxic ADSC-Exo group had significantly more CD31-positive cells than the ADSC-Exo group. In vitro study showed that hypoxic ADSC-Exos treatment significantly increased the migration (at 12 and 24 h) and in vitro capillary network formation (at 12 h) in the human umbilical vein endothelial cells (HUVECs) as compared with the ADSC-Exo group and control group (all P < 0.05). CONCLUSIONS: Co-transplantation of ADSC-Exos can effectively promote the survival of graft, neovascularization and attenuated inflammation in the fat grafts. Hypoxia treatment can further enhance the beneficial effect of ADSC-Exos.


Subject(s)
Adipose Tissue/growth & development , Adipose Tissue/transplantation , Exosomes/transplantation , Graft Survival/physiology , Ischemic Preconditioning/methods , Mesenchymal Stem Cells/ultrastructure , Neovascularization, Physiologic/physiology , Adipose Tissue/cytology , Animals , Exosomes/ultrastructure , Female , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred BALB C , Mice, Nude
8.
Artif Cells Nanomed Biotechnol ; 44(1): 48-55, 2016.
Article in English | MEDLINE | ID: mdl-24697727

ABSTRACT

OBJECTIVE: To examine the effects of sodium pyruvate (SP) on metabolic acidosis. METHODS: For the in vivo experiments, we evaluated effects of SP on an ammonium chloride (NH4Cl)-induced hyperchloremic acidosis rat model. SP was infused at overall doses of 2, 4, and 6 mmol·kg(- 1) for the SP1, SP2, and SP3 groups, respectively. Treatment with sodium bicarbonate (SB) was used as a positive control (2 mmol·kg(- 1)), and treatment with normal saline (NS) was used as a volume control (2 mL·kg(- 1)). Blood was sampled from the ophthalmic venous plexus for pH, blood gases, electrolytes, glucose, creatinine (Cr), and urea analysis after injection. For the in vitro experiment, propionate was applied to induce intracellular acidosis in human endothelial cells. Intracellular pH (pHi) was fluorimetrically measured after the addition of SP. RESULTS: In the in vivo study, the pH of SP1 group showed no significant difference compared with that of the NS group. The SP2 and SP3 groups had a higher pH than the NS group (P < 0.01). The SP3 group had a higher pH than the SB group (P < 0.05) and SP1 group (P < 0.05). Moreover, SP treatment ameliorated the abnormality of calcium and decreased the blood potassium levels. The SP3 group had higher glucose levels than SP1 group (P < 0.05). No significant differences were observed between all the groups in the plasma Cr and urea levels. In the in vitro study, the pHi increased immediately after the addition of SP. CONCLUSION: The data suggest that intravascular treatment with SP represents a novel therapeutic strategy to ameliorate metabolic acidosis.


Subject(s)
Acidosis/blood , Acidosis/drug therapy , Endothelial Cells/drug effects , Pyruvic Acid/pharmacology , Acid-Base Equilibrium/drug effects , Acidosis/chemically induced , Acidosis/pathology , Ammonium Chloride , Animals , Blood Gas Analysis , Blood Glucose/metabolism , Calcium/blood , Cell Line , Creatinine/blood , Endothelial Cells/metabolism , Humans , Hydrogen-Ion Concentration , Male , Potassium/blood , Rats , Rats, Wistar , Sodium/blood , Sodium Bicarbonate/pharmacology , Urea/blood
9.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(5): 667-72, 2015 Sep.
Article in Chinese | MEDLINE | ID: mdl-26619532

ABSTRACT

OBJECTIVE: To isolate and steadily culture kidney stem cells (KSCs) from rat renal papilla, and to identify the biological characteristics of KSCs. METHODS: KSCs were isolated from the tips of renal papilla in 4 weeks-old Sprague-Dawley rats. The morphology of KSCs was observed under inversion microscope, and the phenotye characteristics of kSCs were identified through flow cytometry and immunofluorescence. The abilities of KSCs in adipogenic and osteogenic differentiation were evaluated. The differences of gene expression between KSCs and rat renal tubular epithelial cells (RTECs)were compared using quantitative real time polymerase chain reaction (qRT-PCR). RESULTS: KSCs showed a spindle-shaped and arborization-like growth pattern. Immunofluorescence indicated that KSCs staining with alpha-sooth muscle actin (α-SMA), Vimentin, N-Cadherin, Nestin, CD133 marker, and without E-cadherin, cytokeratin-18 (CK-18), zona occludens protein-1 (ZO-1). The positive staining of CD29, CD90, CD73 were 99. 0%, 95. 8%, 99. 9% respectively, the positive staining of CD45 was 3. 4%. The positive stainings of stem cell marker CD133 and Nestin were 33. 2% and 70. 2% respectively, while the double staining rate was 31. 4%., KSCs showed positive staining by oil red 0 after adipogenic differentiation, and orange calcium deposition by alizarin red staining after osteogenic differentiation. qRT-PCR showed that the expressions of embryonic stem cell marker Nanog, Oct4/pou5f1,Sox2/sry-box-2 in KSCs were higher than those in RTECs (P< 0.01), and the expressions of mesenchymal marker c-SMA, Vimentin were also higher in KSCs (P<0. 01). Compared with RTECs, the expressions of mature epithelium marker E-Cadherin, CK18 in KSCs were lower (P< 0. 01). CONCLUSION: KSCs were isolated successfully and steadily cultured from the rat renal papilla, which were identified with featured biological characteristics.


Subject(s)
Kidney/cytology , Stem Cells/cytology , Adipogenesis , Animals , Cell Culture Techniques , Cell Differentiation , Cell Separation , Cells, Cultured , Flow Cytometry , Osteogenesis , Rats , Rats, Sprague-Dawley
10.
Hepatology ; 62(3): 801-15, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25953743

ABSTRACT

UNLABELLED: Emerging evidence suggests that epithelial-mesenchymal transitions (EMTs) play important roles in tumor metastasis and recurrence. Understanding molecular mechanisms that regulate the EMT process is crucial for improving treatment of hepatocellular carcinoma (HCC). MicroRNAs (miRNAs) play important roles in HCC; however, the mechanisms by which miRNAs target the EMT and their therapeutic potential remains largely unknown. To better explore the roles of miRNAs in the EMT process, we established an EMT model in HCC cells by transforming growth factor beta 1 treatment and found that several tumor-related miRNAs were significantly decreased. Among these miRNAs, miR-125b expression was most strongly suppressed. We also found down-regulation of miR-125b in most HCC cells and clinical specimens, which correlated with cellular differentiation in HCC patients. We then demonstrated that miR-125b overexpression attenuated EMT phenotype in HCC cancer cells, whereas knockdown of miR-125b promoted the EMT phenotype in vitro and in vivo. Moreover, we found that miR-125b attenuated EMT-associated traits, including chemoresistance, migration, and stemness in HCC cells, and negatively correlated with EMT and cancer stem cell (CSC) marker expressions in HCC specimens. miR-125b overexpression could inhibit CSC generation and decrease tumor incidence in the mouse xenograft model. Mechanistically, our data revealed that miR-125b suppressed EMT and EMT-associated traits of HCC cells by targeting small mothers against decapentaplegic (SMAD)2 and 4. Most important, the therapeutic delivery of synthetic miR-125b mimics decreased the target molecule of CSC and inhibited metastasis in the mice model. These findings suggest a potential therapeutic treatment of miR-125b for liver cancer. CONCLUSION: miR-125b exerts inhibitory effects on EMT and EMT-associated traits in HCC by SMAD2 and 4. Ectopic expression of miR-125b provides a promising strategy to treat HCC.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Smad2 Protein/metabolism , Smad4 Protein/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Down-Regulation , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Random Allocation , Sensitivity and Specificity , Transfection , Tumor Cells, Cultured
11.
Zhonghua Xue Ye Xue Za Zhi ; 34(10): 887-92, 2013 Oct.
Article in Chinese | MEDLINE | ID: mdl-24171965

ABSTRACT

OBJECTIVE: To compare the differentiation ability difference of hematopoietic, mesenchymal and endothelial potential between CD41⁺ cells derived from the mouse aorta-gonadmesonephros (AGM) region, yolk sac (YS) and embryonic circulating blood (CB). METHODS: CD41⁺ cells were sorted from AGM, YS and CB. The CD45 and c-kit expression were studied in CD41⁺ cells by flow cytometry. IL-3 and bone morphogenetic protein 4 (BMP-4) treatment together with semi solid culture were used to assess hematopoietic potential difference of CD41⁺ cells. Immunofluorescence staining of α-SMA was used to assess mesenchymal potential difference. The endothelial cell induction system was used to assess endothelial potential difference. RESULTS: The proportions of CD45+ cells in CD41⁺ population were 51.9% (AGM), 45.8% (YS) and 22.2% (CB), respectively, while those of c-kit⁺ cells were 40.0% (AGM), 39.6% (YS) and 36.2% (CB), respectively. After stimulated by IL-3 factor, the number of total colonies increased in all three groups-derived CD41⁺ cells compared to that of unstimulated group[(14.1±1.9) vs (1.2±0.2), (32.4±1.1) vs (18.4±2.2) and (41.8±0.9) vs (10.4±1.8)], (P<0.01). After stimulated by BMP-4 factor, compared to unstimulated group, CFU-Mix colony number in CD41⁺ cells from AGM region and YS were significantly decreased[(0.5±0.6) vs (3.2±0.8), (1.3±0.7) vs (7.4±1.7)](P<0.01), but there was no difference in CB group[(2.5±0.5) vs (3.9±1.5)](P>0.01). The mesenchymal marker α-SMA was highly expressed in CD41⁺ cells from AGM region and YS, but lowly expressed in CD41⁺ cells from CB. CONCLUSION: There are some differences between CD41⁺ cells in AGM region, YS and CB on hematopoietic cell surface marker expression, hematopoietic colony formation with IL-3 and BMP-4 stimulation.


Subject(s)
Aorta/cytology , Cell Differentiation , Gonads/cytology , Mesonephros/cytology , Yolk Sac/cytology , Animals , Bone Morphogenetic Protein 4/pharmacology , Interleukin-3/pharmacology , Mice , Platelet Membrane Glycoprotein IIb/metabolism , Proto-Oncogene Proteins c-kit/metabolism
12.
Zhonghua Yi Xue Za Zhi ; 93(26): 2020-4, 2013 Jul 09.
Article in Chinese | MEDLINE | ID: mdl-24169277

ABSTRACT

OBJECTIVE: To explore the expression of Foxa2 in different pathological types of gastric polyps and examine the correlation with cancerous risk. METHODS: According to computerize random number, a total of 2000 patients were selected to receive endoscopic biopsy during November 2011 to October 2012. Tissues were harvested from 170 with gastric polyps and suspicious cancerous lesions and their histological types detected. There were hyperplastic polyps(n = 35), adenomatous polyps(n = 31), fundic gland polyps(n = 42), advanced gastric cancer tissues (n = 32)and normal gastric mucosa tissues (n = 30). ABC immunohistochemical staining and reverse transcription(RT)-PCR were employed to detect the expression of Foxa2 in these different types of tissues. Imagepro plus was used for quantitative and statistical analyses. RESULTS: A low-level expression of Foxa2 was 3.6% ± 1.3% in normal gastric mucosa group. And its expreesion gradually higher in proliferative inflammatory polyp group(33.1% ± 8.0%), adenomatous polyp group (71.4% ± 1.7%) and gastric cancer group(96.3% ± 0.9%, all P < 0.05). Its expression was 35.6% ± 5.6% in fundic gland polyps, similar to that of proliferative inflammatory polyp group (P > 0.05), it was markedly lower than the gastric cancer group (P < 0.05) and higher than the normal gastric mucosa group (P < 0.05). Correlation analyses of clinicopathological parameters showed that no significant correlation existed between its expression and patient gender, age, predilection, Helicobacter. pylori infection or proton pump inhibitor used (all P > 0.05). However, the size of polyps was correlated with Foxa2 (rs = 0.69, P < 0.05). CONCLUSION: The expression level of Foxa2 in different types of gastric polyps may be used as a clinical predicator of polyps risk.


Subject(s)
Adenomatous Polyps/metabolism , Adenomatous Polyps/pathology , Hepatocyte Nuclear Factor 3-beta/metabolism , Stomach Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/metabolism , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Risk Factors , Stomach Neoplasms/metabolism
13.
Hepatology ; 57(6): 2274-86, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23316018

ABSTRACT

UNLABELLED: Cancer-associated mesenchymal stem cells (MSCs) play a pivotal role in modulating tumor progression. However, the interactions between liver cancer-associated MSCs (LC-MSCs) and hepatocellular carcinoma (HCC) remain unreported. Here, we identified the presence of MSCs in HCC tissues. We also showed that LC-MSCs significantly enhanced tumor growth in vivo and promoted tumor sphere formation in vitro. LC-MSCs also promoted HCC metastasis in an orthotopic liver transplantation model. Complementary DNA (cDNA) microarray analysis showed that S100A4 expression was significantly higher in LC-MSCs compared with liver normal MSCs (LN-MSCs) from adjacent cancer-free tissues. Importantly, the inhibition of S100A4 led to a reduction of proliferation and invasion of HCC cells, while exogenous S100A4 expression in HCC cells resulted in heavier tumors and more metastasis sites. Our results indicate that S100A4 secreted from LC-MSCs can promote HCC cell proliferation and invasion. We then found the expression of oncogenic microRNA (miR)-155 in HCC cells was significantly up-regulated by coculture with LC-MSCs and by S100A4 ectopic overexpression. The invasion-promoting effects of S100A4 were significantly attenuated by a miR-155 inhibitor. These results suggest that S100A4 exerts its effects through the regulation of miR-155 expression in HCC cells. We demonstrate that S100A4 secreted from LC-MSCs promotes the expression of miR-155, which mediates the down-regulation of suppressor of cytokine signaling 1, leading to the subsequent activation of STAT3 signaling. This promotes the expression of matrix metalloproteinases 9, which results in increased tumor invasiveness. CONCLUSION: S100A4 secreted from LC-MSCs is involved in the modulation of HCC progression, and may be a potential therapeutic target. (HEPATOLOGY 2013).


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , S100 Proteins/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Disease Progression , Humans , Liver Neoplasms/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , S100 Calcium-Binding Protein A4 , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
14.
Cell Reprogram ; 14(1): 88-97, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22313114

ABSTRACT

Blood cells transfusion and hematopoietic stem cells (HSCs) transplantation are important methods for cell therapy. They are widely used in the treatment of incurable hematological disorder, infectious diseases, genetic diseases, and immunologic deficiency. However, their availability is limited by quantity, capacity of proliferation and the risk of blood transfusion complications. Recently, human embryonic stem cells (hESCs) have been shown to be an alternative resource for the generation of hematopoietic cells. In the current study, we describe a novel method for the efficient production of hematopoietic cells from hESCs. The stable human fetal liver stromal cell lines (hFLSCs) expressing erythropoietin (EPO) were established using the lentiviral system. We observed that the supernatant from the EPO transfected hFLSCs could induce the hESCs differentiation into hematopoietic cells, especially erythroid cells. They not only expressed fetal and embryonic globins but also expressed the adult-globin chain on further maturation. In addition, these hESCs-derived erythroid cells possess oxygen-transporting capacity, which indicated hESCs could generate terminally mature progenies. This should be useful for ultimately developing an animal-free culture system to generate large numbers of erythroid cells from hESCs and provide an experimental model to study early human erythropoiesis.


Subject(s)
Embryonic Stem Cells/cytology , Erythropoietin/metabolism , Fetus/cytology , Hematopoiesis/physiology , Liver/cytology , Stromal Cells/cytology , Stromal Cells/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Culture Media, Conditioned/pharmacology , Embryonic Stem Cells/physiology , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythropoietin/genetics , Fetal Globulins/metabolism , Humans , Lentivirus/genetics , Liver/embryology , Liver/metabolism , Transfection
15.
Mol Cancer Res ; 10(3): 326-35, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22258766

ABSTRACT

SPINDLIN1, a new member of the SPIN/SSTY gene family, was first identified as a gene highly expressed in ovarian cancer cells. We have previously shown that it is involved in the process of spindle organization and chromosomal stability and plays a role in the development of cancer. Nevertheless, the mechanisms underlying its oncogenic role are still largely unknown. Here, we first showed that expression of SPINDLIN1 is upregulated in clinical tumors. Ectopic expression of SPINDLIN1 promoted cancer cell proliferation and activated WNT/T-cell factor (TCF)-4 signaling. The Ser84 and Ser99 amino acids within SPINDLIN1 were further identified as the key functional sites in WNT/TCF-4 signaling activation. Mutation of these two sites of SPINDLIN1 abolished its effects on promoting WNT/TCF-4 signaling and cancer cell proliferation. We further found that Aurora-A could interact with and phosphorylate SPINDLIN1 at its key functional sites, Ser84 and Ser99, suggesting that phosphorylation of SPINDLIN1 is involved in its oncogenic function. Collectively, these results suggest that SPINDLIN1, which may be a novel substrate of the Aurora-A kinase, promotes cancer cell growth through WNT/TCF-4 signaling activation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasms/pathology , Phosphoproteins/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , Aurora Kinases , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mutation/genetics , Neoplasm Invasiveness , Neoplasms/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Transcription Factor 4
16.
J Mol Med (Berl) ; 90(4): 389-400, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22038097

ABSTRACT

Human mesenchymal stem cells (MSCs) have therapeutic potential because of their ability to self-renew and differentiate into multiple tissues. However, senescence often occurs in MSCs when they are cultured in vitro and the molecular mechanisms underlying this effect remain unclear. In this study, we found that NAD-dependent protein deacetylase SIRT1 is differentially expressed in both human bone marrow-derived MSCs (B-MSCs) and adipose tissue-derived MSCs after increasing passages of cell culture. Using lentiviral shRNA we demonstrated that selective knockdown of SIRT1 in human MSCs at early passage slows down cell growth and accelerates cellular senescence. Conversely, overexpression of SIRT1 delays senescence in B-MSCs that have undergone prolonged in vitro culturing and the cells do not lose adipogenic and osteogenic potential. In addition, we found that the delayed accumulation of the protein p16 is involved in the effect of SIRT1. However, resveratrol, which has been used as an activator of SIRT1 deacetylase activity, only transiently promotes proliferation of B-MSCs. Our findings will help us understand the role of SIRT1 in the aging of normal diploid cells and may contribute to the prevention of human MSCs senescence thus benefiting MSCs-based tissue engineering and therapies.


Subject(s)
Mesenchymal Stem Cells/cytology , Sirtuin 1/metabolism , Adipose Tissue/cytology , Adult , Bone Marrow Cells/cytology , Cell Differentiation , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Gene Knockdown Techniques , Humans , Male , Mesenchymal Stem Cells/metabolism , Resveratrol , Sirtuin 1/genetics , Stilbenes/pharmacology , Up-Regulation , Young Adult
17.
Breast Cancer Res Treat ; 132(1): 153-64, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21584665

ABSTRACT

Mesenchymal stem cells (MSCs) play a critical role in promoting cancer progression. However, it is not clear whether MSCs are located in breast cancer tissues and correlated with tumor proliferation. The aim of this study was to investigate the presence of MSCs in breast cancer tissues and evaluate their interactions with cancer cells. We successfully isolated and identified MSCs from primary breast cancer tissues. Breast cancer-associated MSCs (BC-MSCs) showed homogenous immunophenotype, and possessed tri-lineage differentiation potential (osteoblast, adipocyte, and chondrocyte). When co-transplanted with cancer cells in a xenograft model in vivo, BC-MSCs significantly increased the volume and weight of tumors. We observed that BC-MSCs stimulated mammosphere formation in the transwell co-culture system in vitro. This effect was significantly suppressed by the EGF receptor inhibitor. We verified that BC-MSCs could secrete EGF and activate cancer cell's EGF receptors. Furthermore, our data showed that EGF derived from BC-MSCs could promote mammosphere formation via the PI3K/Akt signaling pathway. Our results confirmed the presence of MSC in primary breast cancer tissues, and they could provide a favorable microenvironment for tumor cell growth in vivo, partially enhance mammosphere formation via the EGF/EGFR/Akt pathway.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Epidermal Growth Factor/physiology , Mesenchymal Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Spheroids, Cellular/metabolism , Animals , Antigens, Differentiation/metabolism , Cell Differentiation , Cell Proliferation , Cell Shape , Coculture Techniques , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tumor Burden , Tumor Cells, Cultured
18.
Hepatology ; 54(5): 1808-18, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22045676

ABSTRACT

UNLABELLED: The high incidence rate of hepatocellular carcinoma (HCC) is mainly the result of frequent metastasis and tumor recurrence. Unfortunately, the underlying molecular mechanisms driving HCC metastasis are still not fully understood. It has been demonstrated that tumor stroma cells contribute to primary tumor growth and metastasis. Within the HCC environment, activated hepatic stellate cells (HSCs) can release a number of molecules and enhance cancer cell proliferation and invasiveness in a paracrine manner. Here, for the first time, we demonstrate that epimorphin (EPM; also called syntaxin-2), an extracellular protein, is strongly elevated in activated HSCs within tumor stroma. We show that knockdown of EPM expression in HSCs substantially abolishes their effects on cancer cell invasion and metastasis. Ectopic expression of EPM in HCC cancer cells enhances their invasiveness; we demonstrate that the cells expressing EPM have markedly increased metastasis potential. Furthermore, EPM-mediated invasion and metastasis of cancer cells is found to require up-regulation of matrix metalloproteinase-9 (MMP-9) through the activation of focal adhesion kinase (FAK)/extracellular signal-regulated kinase (ERK) axis. CONCLUSION: Our results show that EPM, secreted by activated HSCs within HCC stroma, promotes invasion and metastasis of cancer cells by activating MMP-9 expression through the FAK-ERK pathway.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Signaling System/physiology , Syntaxin 1/metabolism , Cell Division/physiology , Cell Movement/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Hep G2 Cells , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism , Liver/pathology , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness
19.
In Vitro Cell Dev Biol Anim ; 47(9): 601-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21938589

ABSTRACT

Although human amniotic fluid is an attractive source of multipotent stem cells, the potential of amniotic fluid stem cells (AFSCs) to differentiate into hepatic cells has not been extensively evaluated. In this study, we examined whether human AFSCs can differentiate into a hepatic cell lineage in vitro and in vivo. After being treated with cytokines (fibroblast growth factor 4, basic fibroblast growth factor, hepatocyte growth factor, and oncostatin), AFSCs developed a morphology similar to that of hepatocytes. RT-PCR and immunofluorescence analysis showed that the treated AFSCs expressed the hepatocyte-specific markers albumin, cytokeratin 18, and alpha-fetoprotein. The differentiated cells also developed hepatocyte-specific functions, i.e., they secreted albumin, absorbed indocyanine green, and stored glycogen. When transplanted into CCl(4)-injured immunodeficient mice, undifferentiated AFSCs were integrated into the liver tissue, and they expressed markers characteristic of mature human hepatocytes. Although integration of AFSCs into the liver was limited (0.1-0.3% of hepatocytes), histological analysis showed that the recipient mice recovered more rapidly from CCl(4) injury than CCl(4)-injured mice that did not receive AFSCs. AFSCs can differentiate into hepatocyte-like cells in vitro and in vivo and can represent an easily accessible source of progenitor cells for hepatocyte regeneration and liver cell transplantation.


Subject(s)
Amniotic Fluid/cytology , Cell Differentiation , Hepatocytes/cytology , Stem Cells/cytology , Animals , Biological Assay , Carbon Tetrachloride , Cell Shape , Fluorescent Antibody Technique , Gene Expression Regulation , Hepatocytes/metabolism , Humans , Mice , Mice, Nude , Organ Specificity/genetics , Stem Cell Transplantation
20.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 33(4): 402-7, 2011 Aug.
Article in Chinese | MEDLINE | ID: mdl-21906449

ABSTRACT

OBJECTIVE: To explore the method of constructing tissue-engineered skin using melanocytes and bone marrow mesenchymal stem cells (BMSCs) in vivo. METHODS: Melanocytes were isolated from human foreskin. BMSCs were isolated from human bone marrow. Both of them were co-cultured at a ratio of 1:10, and then were implanted into the collagen membrane to construct the tissue-engineered skin, which was applied for wound repair in nude mice. The effectiveness of wound repair and the distribution of melanocytes were evaluated by morphological observation, in vivo 4,6-diamidino-2-phenylindole, dihydrochloride (DAPI) fluorescent staining tracing, HE staining, S-100 immunohistochemistry, and transmission electron microscopy. RESULTS: The wounds were satisfactorily repaired among the nude mice. The melanocytes were distributed in the skin with normal structure, as confirmed by DAPI fluorescent staining tracing, HE staining, S-100 immunohistochemistry, and transmission electron microscopy. CONCLUSION: Melanocytes and BMSCs, after proper in vitro culture at an appropriate ratio, can construct the tissue-engineered skin with I type collagen membrane.


Subject(s)
Bone Marrow Cells/cytology , Melanocytes/cytology , Mesenchymal Stem Cells/cytology , Skin, Artificial , Tissue Engineering , Animals , Cells, Cultured , Coculture Techniques , Collagen Type I , Humans , Mice , Mice, Nude , Skin/injuries
SELECTION OF CITATIONS
SEARCH DETAIL
...