Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Life Sci ; 293: 120050, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35026215

ABSTRACT

Gastric cancer (GC) is an aggressive disease with one of the highest mortality rates in the world. In the early stage, most patients are asymptomatic and early diagnosis is difficult. Recently, cancer stem cells (CSCs) have been highlighted as crucial emerging factors in the initiation or invasiveness of solid tumors. CD133, a CSC marker, is highly expressed in various tumors including gastric cancer. CD133-positive cells showed elevated malignant biological behaviors and CD133 upregulation is related to chemoresistance, cancer relapse, and poor prognosis. CD133 also plays an important role in the progression of tumors and metastasis. This review summarizes the current knowledge of the role of CD133 expression in GC and aims to contribute at identifying promising new strategies for treatment and management of gastric cancer.


Subject(s)
AC133 Antigen/biosynthesis , Biomarkers, Tumor/metabolism , Neoplastic Stem Cells/metabolism , Stomach Neoplasms/metabolism , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/pathology , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy
2.
Biomed Pharmacother ; 137: 111364, 2021 May.
Article in English | MEDLINE | ID: mdl-33592546

ABSTRACT

Colorectal cancer (CRC) is considered one of the leading types of cancer in the world. CD133, as a cancer stem cell marker, has a pivotal role in the development of drug resistance, migration, and stemness properties of CRC cells. This study was designed to check the combined effect of CD133 siRNA and Oxaliplatin on proliferation, migration, apoptosis, and stemness properties of CRC cells in the HT-29 cell line. MTT assay was performed to define the combined effect of CD133 siRNA and Oxaliplatin on the viability of HT-29 cells, and it showed that the combination of CD133 siRNA and Oxaliplatin could reduce the IC50 of this drug from 32.85 to 19.75 nmol. In order to figure out the effect of this combination therapy on CD133 expression at the gene and protein level, qRT-PCR and western blot were exploited, respectively. The results demonstrated that the silencing of CD133 could reduce the relative expression of this marker to about 0.00001 compared to the control group and reduce the protein level to 0.01. The ability of cell migration was tested by wound healing assay as well. Also, colony formation and sphere formation were conducted to assess the stemness properties in the combination group. Flow cytometry was conducted to investigate the apoptosis (15%), cell cycle (about 10% arresting in G0-G1 phase), and surface expression of CD133 in different groups (from 39.3% in the control group to 2.41 in the combination group). Finally, the expression of migration-, and stemness-associated genes were measured by qRT-PCR. We indicated that silencing of CD133 reduces the migration and stemness properties of colorectal cancerous cells. This suppression makes HT-29 cells more sensitive to Oxaliplatin and reduces the effective dose of this chemical drug. Therefore, the suppression of CD133 in combination with Oxaliplatin treatment might be a promising therapeutic approach in the treatment of colorectal cancer.


Subject(s)
AC133 Antigen/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Oxaliplatin/therapeutic use , AC133 Antigen/genetics , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Drug Screening Assays, Antitumor , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic , Gene Silencing , HT29 Cells , Humans , Neoplastic Stem Cells/drug effects , RNA, Small Interfering/therapeutic use , Tumor Stem Cell Assay
3.
Cell Biol Int ; 44(2): 368-380, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31579983

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of death worldwide. Recently, the role of cancer stem cells (CSCs) has been highlighted as a crucial emerging factor in chemoresistance, cancer relapse, and metastasis. CD133 is a surface marker of CSCs and has been argued to have prognostic and therapeutic values in CRC along with its related pathways such as Wnt, Notch, and hedgehog. Several studies have successfully applied targeted therapies against CD133 in CRC models namely bispecific antibodies (BiAbs) and anti-Wnt and notch pathways agents. These studies have yielded initial promising results in this regard. However, none of the therapeutics have been used in the clinical setting and their efficacy and adverse effects profile are yet to be elucidated. This review aims to gather the old and most recent data on the prognostic and therapeutic values of CD133 and CD133-targeted therapies in CRC.


Subject(s)
AC133 Antigen/antagonists & inhibitors , AC133 Antigen/metabolism , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Neoplastic Stem Cells/pathology
4.
Clin Cancer Res ; 26(5): 1054-1064, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31732520

ABSTRACT

PURPOSE: Aggressive variant prostate cancer (AVPC) is a nonandrogen receptor-driven form of disease that arises in men in whom standard-of-care therapies have failed. Therapeutic options for AVPC are limited, and the development of novel therapeutics is significantly hindered by the inability to accurately quantify patient response to therapy by imaging. Imaging modalities that accurately and sensitively detect the bone and visceral metastases associated with AVPC do not exist. EXPERIMENTAL DESIGN: This study investigated the transmembrane protein CD133 as a targetable cell surface antigen in AVPC. We evaluated the expression of CD133 by microarray and IHC analysis. The imaging potential of the CD133-targeted IgG (HA10 IgG) was evaluated in preclinical prostate cancer models using two different imaging modalities: near-infrared and PET imaging. RESULTS: Evaluation of the patient data demonstrated that CD133 is overexpressed in a specific phenotype of AVPC that is androgen receptor indifferent and neuroendocrine differentiated. In addition, HA10 IgG was selective for CD133-expressing tumors in all preclinical imaging studies. PET imaging with [89Zr]Zr-HA10 IgG revealed a mean %ID/g of 24.30 ± 3.19 in CD133-positive metastatic lesions as compared with 11.82 ± 0.57 in CD133-negative lesions after 72 hours (P = 0.0069). Ex vivo biodistribution showed similar trends as signals were increased by nearly 3-fold in CD133-positive tumors (P < 0.0001). CONCLUSIONS: To our knowledge, this is the first study to define CD133 as a targetable marker of AVPC. Similarly, we have developed a novel imaging agent, which is selective for CD133-expressing tumors, resulting in a noninvasive PET imaging approach to more effectively detect and monitor AVPC.


Subject(s)
AC133 Antigen/metabolism , Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/metabolism , Molecular Imaging/methods , Positron-Emission Tomography/methods , Prostatic Neoplasms/diagnostic imaging , Radioisotopes/pharmacokinetics , Zirconium/pharmacokinetics , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/immunology , Animals , Biomarkers, Tumor/immunology , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Radiopharmaceuticals/pharmacokinetics , Receptors, Androgen/metabolism , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Biol Chem ; 294(45): 16634-16649, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31533988

ABSTRACT

The existence and regenerative potential of resident stem and progenitor cells in the adult pancreas are controversial topics. A question that has been only minimally addressed is the capacity of a progenitor cell to self-renew, a key attribute that defines stem cells. Previously, our laboratory has identified putative stem and progenitor cells from the adult murine pancreas. Using an ex vivo colony/organoid culture system, we demonstrated that these stem/progenitor-like cells have self-renewal and multilineage differentiation potential. We have named these cells pancreatic colony-forming units (PCFUs) because they can give rise to three-dimensional colonies. However, the molecular mechanisms by which PCFUs self-renew have remained largely unknown. Here, we tested the hypothesis that PCFU self-renewal requires GLIS family zinc finger 3 (GLIS3), a zinc-finger transcription factor important in pancreas development. Pancreata from 2- to 4-month-old mice were dissociated, sorted for CD133highCD71low ductal cells, known to be enriched for PCFUs, and virally transduced with shRNAs to knock down GLIS3 and other proteins. We then plated these cells into our colony assays and analyzed the resulting colonies for protein and gene expression. Our results revealed a previously unknown GLIS3-to-CD133-to-WNT signaling axis in which GLIS3 and CD133 act as factors necessary for maintaining WNT receptors and signaling molecules in colonies, allowing responses to WNT ligands. Additionally, we found that CD133, but not GLIS3 or WNT, is required for phosphoinositide 3-kinase (PI3K)/AKT Ser/Thr kinase (AKT)-mediated PCFU survival. Collectively, our results uncover a molecular pathway that maintains self-renewal of adult murine PCFUs.


Subject(s)
AC133 Antigen/metabolism , DNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Wnt Proteins/metabolism , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Self Renewal , Cell Survival , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Pancreas/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Thrombospondins/genetics , Thrombospondins/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Wnt Proteins/genetics , beta Catenin/metabolism
6.
Eur J Med Chem ; 180: 224-237, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31306909

ABSTRACT

Cytotoxic effects of (R)-4'-methylklavuzon were investigated on hepatocellular carcinoma cells (HuH-7 and HepG2) and HuH-7 EpCAM+/CD133+ cancer stem cells. IC50 of (R)-4'-methylklavuzon was found as 1.25 µM for HuH-7 parental cells while it was found as 2.50 µM for HuH-7 EpCAM+/CD133+ cancer stem cells. (R)-4'-methylklavuzon tended to show more efficient in vitro cytotoxicity with its lower IC50 values on hepatocellular carcinoma cell lines compared to its lead molecule, goniothalamin and FDA-approved drugs, sorafenib and regorafenib. Cell-based Sirtuin/HDAC enzyme activity measurements revealed that endogenous Sirtuin/HDAC enzymes were reduced by 40% compared to control. SIRT1 protein levels were upregulated indicating triggered DNA repair mechanism. p53 was overexpressed in HepG2 cells. (R)-4'-methylklavuzon inhibited CRM1 protein providing increased retention of p53 and RIOK2 protein in the nucleus. HuH-7 parental and EpCAM+/CD133+ cancer stem cell spheroids lost intact morphology. 3D HepG2 spheroid viabilities were decreased in a correlation with upregulation in p53 protein levels.


Subject(s)
AC133 Antigen/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Karyopherins/antagonists & inhibitors , Liver Neoplasms/drug therapy , Naphthalenes/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Sirtuin 1/antagonists & inhibitors , AC133 Antigen/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epithelial Cell Adhesion Molecule/metabolism , Hep G2 Cells , Humans , Karyopherins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Molecular Structure , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Sirtuin 1/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured , Exportin 1 Protein
7.
Chem Biol Interact ; 309: 108713, 2019 Aug 25.
Article in English | MEDLINE | ID: mdl-31226288

ABSTRACT

Liver cancer is one of the most frequently occurring types of cancer with high mortality rate. Hepatocellular carcinoma (HCC) frequently metastasizes to lung, portal vein, and portal lymph nodes and most HCCs show strong resistance to conventional anticancer drugs. Cancer stem cells (CSCs) are considered to be responsible for resistance to therapies. Hence, recent advancements in the use of liver cancer stem cells (LCSCs) are rapidly gaining recognition as an efficient and organized means for developing antitumor agents. We aimed to use a non-target-based high-throughput screening (HTS) approach to specifically target α-fetoprotein (AFP)+/cluster of differentiation (CD)133+ HCC present in mixed populations of HCC cells and hepatocytes. Herein, we identified actinomycin D (ActD) as a potential antitumor agent that significantly inhibits activity of LCSCs without affecting the co-cultured hepatocytes. To determine the mechanism of ActD-induced tumor-specificity in LCSC, we applied various cell-based assay models in vitro. In fact, ActD significantly increased reactive oxygen species (ROS) accumulation and DNA damage in Huh7 HCC cells, but not in Fa2N-4 cells, immortalized hepatocytes. Treatment of spheroid-forming LCSCs with ActD effectively decreased spheroid formation and the CD133+ HCC cell population. Importantly, these ActD-mediated effects are a result of inhibition of cystine/glutamate transporter xCT expression, via attenuation of CD133 synthesis. These results indicate that ActD suppresses stemness and malignant properties in HCC cells through destabilization of xCT, by inhibition of CD133 expression in LCSCs. The effects of ActD on LCSCs provide novel therapeutic strategies for targeting cancer stem-like cells in liver cancer.


Subject(s)
AC133 Antigen/metabolism , Amino Acid Transport System y+/metabolism , Dactinomycin/pharmacology , Gene Expression/drug effects , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/genetics , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage/drug effects , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Spheroids, Cellular/drug effects
8.
Nanotechnology ; 30(35): 355101, 2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31082814

ABSTRACT

Metallic nanorods are promising agents for a wide range of biomedical applications. We report an optical hyperthermia method capable of inducing slowdown tumor progression of an experimental in vivo CT-2A glioblastoma tumor. The tumor model used in this research is based on the transplantation of mouse astrocytoma CT-2A cells in the striatum of mice by intracranial stereotaxic surgery. Two weeks after cell implant, the resulting tumor is treated by irradiating intratumoral injected gold nanorods, biofunctionalized with CD133 antibody (B-GNRs), using a continuous wave laser. Nanoparticles convert the absorbed light into localized heat (reaching up to 44 °C) due to the effect of surface plasmon resonance. A significant slowdown in CT-2A tumor progression is evident, by histology and magnetic resonance imaging, at one (p = 0.03) and two weeks (p = 0.008) after irradiation treatment. A notable deceleration in tumor size (15%-75%) as compared to the control untreated groups, it is observed. Thus, laser irradiation of B-GNRs is found to be effective for the treatment of CT-2A tumor progression. Similarities between the pre-clinical CT-2A tumor model and the human astrocytoma disease, in terms of anatomy, metastatic behavior and histopathology, suggest that hyperthermic treatment by laser irradiation of B-GNRs administered into high-grade human astrocytoma might constitute a promising alternative treatment to limit the progression of this deadly disease.


Subject(s)
Astrocytoma/therapy , Brain Neoplasms/therapy , Gold/pharmacology , Hyperthermia, Induced/methods , Laser Therapy/methods , Nanotubes/chemistry , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/immunology , Animals , Antibodies, Neutralizing/pharmacology , Astrocytoma/immunology , Astrocytoma/pathology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Gold/administration & dosage , Gold/chemistry , Humans , Injections, Intralesional , Lasers , Mice , Mice, Inbred C57BL , Nanotubes/ultrastructure , Neoplasm Transplantation , Stereotaxic Techniques , Surface Plasmon Resonance , Tumor Burden/radiation effects
9.
Hum Gene Ther ; 30(4): 446-458, 2019 04.
Article in English | MEDLINE | ID: mdl-29706119

ABSTRACT

CRISPR/Cas9-mediated programmed cell death protein 1 (PD-1) disruption in chimeric antigen receptor (CAR) T cells could be an appealing choice to improve the therapeutic efficacy of CAR T cells in an immunosuppressive tumor microenvironment. In most of the reported cases, Cas9 was delivered into T cells by way of electroporation with RNA or protein. However, transient expression of Cas9 by transfection with a plasmid encoding its gene is apparently simpler, as it avoids the steps of in vitro transcription of DNA or protein production. This study tried nucleofection into human primary T cells of plasmids encoding both CRISPR/Cas9 for disrupting the PD-1 gene and the piggyBac transposon system for expressing CD133-specific CAR in one reaction. Based on drug selection, CD133-specific CAR T cells were obtained in which, on average, 91.5% of the PD-1 gene sites were disrupted, but almost no Cas9 gene expression was found in the final engineered CAR T cells. The PD-1-deficient CD133-specific CAR T cells showed similar levels of cytokine secretion and improved proliferation and cytotoxicity in vitro, and enhanced inhibition of tumor growth in an orthotopic mouse model of glioma, compared to conventional CD133-CAR T cells. The described method could be useful for the production of PD-1-deficient CAR T cells for cancer immunotherapy.


Subject(s)
AC133 Antigen/immunology , CRISPR-Cas Systems , Gene Silencing , Plasmids/genetics , Programmed Cell Death 1 Receptor/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , AC133 Antigen/antagonists & inhibitors , Animals , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Gene Editing , Gene Knockdown Techniques , Genetic Engineering , Humans , Immunophenotyping , Immunotherapy, Adoptive , Lymphocyte Activation , Mice , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/metabolism , Receptors, Chimeric Antigen/genetics , T-Cell Antigen Receptor Specificity , Transfection/methods , Xenograft Model Antitumor Assays
10.
Int J Oncol ; 53(3): 1289-1300, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30015824

ABSTRACT

As the leading cause of cancer-associated mortality worldwide, lung cancer is often associated with therapy failure and decreases in survival; these factors are often attributed to lung cancer-initiating cells (CICs). In addition, sufficient evidence has suggested that simultaneous targeting of CICs, together with cancer cells, is critical for the achievement of preferable therapeutic efficacy, due to the spontaneous conversion between CICs and cancer cells. Salinomycin sodium (SS) is an antibacterial therapeutic agent that exerts potent activity against CICs in various types of cancer, including lung cancer. The present study generated SS lipid-polymer hybrid nanoparticles (NPs) with cluster of differentiation (CD)133 and epidermal growth factor receptor (EGFR) antibodies (CD133/EGFR SS NPs) for the simultaneous treatment of lung CICs and cancer cells. The activity of CD133/EGFR SS NPs was analyzed using cytotoxicity and tumorsphere formation assays, flow cytometry, and an in vivo anticancer assay in mice bearing lung cancer xenografts. The results revealed that CD133/EGFR SS NPs effectively promoted SS delivery to lung CICs and cancer cells, achieving superior therapeutic effects compared with non-targeted NPs or NPs with a single antibody. Furthermore, CD133/EGFR SS NPs exhibited the best efficacy in inhibiting tumor growth compared with the control agents in lung cancer-bearing mice. In conclusion, CD133/EGFR SS NPs may be capable of efficiently targeting and treating lung CICs together with cancer cells, and may represent an effective treatment for lung cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Drug Delivery Systems/methods , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Pyrans/pharmacology , AC133 Antigen/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Humans , Lactic Acid/chemistry , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Pyrans/therapeutic use , Xenograft Model Antitumor Assays
11.
Blood Adv ; 2(11): 1250-1258, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29858209

ABSTRACT

Acute myeloid leukemia (AML), the most common acute leukemia in adults and the second most common cancer in children, is still a lethal disease in the majority of patients, but immunologic approaches have improved outcome. Bispecific antibodies (BsAbs) are novel immunotherapeutics that can redirect immune cells against AML. We now report a tetravalent (2+2) humanized BsAb in the immunoglobulin G light chain single chain fragment variable [IgG(L)-scFv] format to engage polyclonal T cells to kill CD33+ AML targets. In vitro, this BsAb demonstrated strong antigen-specific T-cell-dependent cell-mediated cytotoxicity (TDCC) with an 50% effective concentration (EC50) in the femtomolar range that translated into treatment of established human AML IV xenografts in vivo. Importantly, it could redirect intraperitoneally injected T cells to ablate established and rapidly growing extramedullary subcutaneous AML xenografts in vivo. Furthermore, internalization of CD33 upon BsAb binding was identical to that of a bivalent (1+1) heterodimer, both being substantially less than anti-CD33 IgG. In contrast to the heterodimer, the tetravalent IgG-scFv BsAb was >10-fold more efficient in TDCC of AML cells in vitro and in vivo. This BsAb did not react with and did not kill CD38-CD34+ hematopoietic stem cells from cord blood. We conclude that the novel anti-CD33 IgG(L)-scFv BsAb construct reported here is a potential candidate for clinical development.


Subject(s)
AC133 Antigen , Antibodies, Bispecific/pharmacology , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological/pharmacology , Leukemia, Myeloid, Acute , T-Lymphocytes/immunology , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/immunology , Animals , Cell Line, Tumor , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/immunology , Mice , Mice, Knockout , Mice, SCID , Xenograft Model Antitumor Assays
12.
Sci Rep ; 8(1): 5674, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29618746

ABSTRACT

This study is to explore the molecular regulation mechanism of CD133 which is associated with malignancy and poor prognosis of blood system diseases. CD133+HUCB-MNC (human umbilical cord blood mononuclear cells) and CD133-HUCB-MNC were isolated and amplificated from umbilical cord blood, and then were exposed to different doses of radiation and subjected to a clonogenic assay. CCK-8 kit was used to detect cell viability, Annexin V-FITC/PI cell apoptosis detection kit was used for the detection of apoptotic cells and the BrdU assay was performed by flow cytometry. The expression of protein was analyzed by western blots. The profile of miRNA expression in response to radiation was examined and validated by RT-PCR. miR-142-3p inhibited the expression of CD133 in umbilical cord blood mononuclear cells to increase radiosensitivity. CD133+HUCB-MNC cells were more radioresistant compared with CD133-HUCB-MNC cells. CD133+HUCB-MNC cells showed higher p-AKT and p-ERK levels after radiation. And miR-142-3p acted on 3'UTR of CD133 mRNA to inhibit CD133 expression. Moreover, miRNA-142-3p mimic increased radiosensitivity in CD133+HUCB-MNC cells. Our results elucidated a novel regulation pathway in hematopoietic stem cells and suggested a potential therapeutic approach for blood system diseases therapy.


Subject(s)
AC133 Antigen/antagonists & inhibitors , Fetal Blood/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Leukocytes, Mononuclear/radiation effects , MicroRNAs/genetics , Radiation Tolerance , AC133 Antigen/genetics , AC133 Antigen/metabolism , Apoptosis , Cell Cycle , Cell Proliferation , Cells, Cultured , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
13.
Cell Commun Signal ; 16(1): 9, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29530069

ABSTRACT

BACKGROUND: The WNT-beta-catenin pathway is known to regulate cellular homeostasis during development and tissue regeneration. Activation of WNT signaling increases the stability of cytoplasmic beta-catenin and enhances its nuclear translocation. Nuclear beta-catenin function is regulated by transcriptional co-factors such as CREB binding protein (CBP) and p300. Hyper-activated WNT-beta-catenin signaling is associated with many cancers. However, its role in inducing stemness to liver cancer cells, its autoregulation and how it regulates tumor suppressor pathways are not well understood. Here we have investigated the role of CBP-beta-catenin signaling on the expression of CD133, a known stem cell antigen and PP2A-PTEN pathway in tumor initiating liver cancer cells. METHODS: Human hepatoblastoma cell line HepG2 and clonally expanded CD133 expressing tumor initiating liver cells (TICs) from premalignant murine liver were used in this study. CBP-beta-catenin inhibitor ICG001 was used to target CBP-beta catenin signaling in liver cancer cells in vitro. Western blotting and real time PCR (qPCR) were used to quantify protein expression/phosphorylation and mRNA levels, respectively. CBP and CD133 gene silencing was performed by siRNA transfection. Fluorescence Activated Cell Sorting (FACS) was performed to quantify CD133 positive cells. Protein Phosphatase (PP2A) activity was measured after PP2AC immunoprecipitation. RESULTS: CBP inhibitor ICG001 and CBP silencing significantly reduced CD133 expression and anchorage independent growth in HepG2 and murine TICs. CD133 silencing in TICs decreased cell proliferation and expression levels of cell cycle regulatory genes, CyclinD1 and CyclinA2. ICG001 treatment and CBP silencing reduced the levels of phosphoSer380/Tyr382/383PTEN, phosphoSer473-AKT, Phospho-Ser552beta-catenin in TICs. ICG001 mediated de-phosphorylation of PTEN in TICs was PP2A dependent and partly prevented by co-treatment with PP2A inhibitor okadaic acid. CONCLUSIONS: CBP-beta-catenin signaling promotes stemness via CD133 induction and cell proliferation in TICs. We found a novel functional link between CBP-beta-catenin and PP2A-PTEN-AKT pathway in liver TICs. Therefore, CBP-beta-catenin-PP2A-PTEN-AKT signaling axis could be a novel therapeutic target to prevent liver tumor initiation and cancer recurrence.


Subject(s)
AC133 Antigen/metabolism , CREB-Binding Protein/metabolism , Carboxylic Ester Hydrolases/metabolism , PTEN Phosphohydrolase/metabolism , Signal Transduction , beta Catenin/metabolism , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/genetics , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CREB-Binding Protein/antagonists & inhibitors , CREB-Binding Protein/genetics , Cell Line, Tumor , Cell Proliferation , Cyclin A2/metabolism , Cyclin D1/metabolism , Down-Regulation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidinones/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , beta Catenin/antagonists & inhibitors
14.
World J Surg Oncol ; 15(1): 167, 2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28854941

ABSTRACT

BACKGROUND: Lung cancer stem cells (CSCs) are considered to be the seed of lung cancer, and CD133 is a marker of lung CSCs. Here, we developed gefitinib-loaded poly(ethylene glycol) 2000-distearoylphosphatidylethanolamine nanomicelles with CD133 aptamers (M-Gef-CD133) to eliminate CD133+ lung CSCs. METHODS: M-Gef-CD133 was prepared using a lipid-film-based approach. The targeting and activity of M-Gef-CD133 towards lung CSCs were evaluated. RESULTS: M-Gef-CD133 were small (25 nm) and showed enhanced cytotoxic effect towards CD133+ lung CSCs compared with non-targeted M-Gef and gefitinib. Notably, M-Gef-CD133 could significantly reduce tumor sphere formation and the percentage of CD133+ lung CSCs, indicating that it possesses selective toxicity against CD133+ lung CSCs. CONCLUSIONS: The interaction of CD133 aptamers and CD133 shows promise in the delivery of gefitinib to CD133+ lung CSCs, and M-Gef-CD133 represents a promising treatment to target lung CSCs.


Subject(s)
AC133 Antigen/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Quinazolines/pharmacology , A549 Cells , AC133 Antigen/metabolism , Animals , Antineoplastic Agents/therapeutic use , Aptamers, Peptide/chemistry , Gefitinib , Humans , Mice , Mice, SCID , Nanoparticles/chemistry , Neoplastic Stem Cells/metabolism , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Quinazolines/therapeutic use , Xenograft Model Antitumor Assays
15.
Hum Gene Ther ; 28(10): 886-896, 2017 10.
Article in English | MEDLINE | ID: mdl-28836469

ABSTRACT

Ovarian cancer represents the most lethal gynecological cancer. Although cytoreductive chemotherapy and surgery lead to complete macroscopic tumor removal, most of the patients in advanced stages suffer from recurrent disease and subsequently die. This may be explained by the activity of cancer stem cells (CSC), which are a subpopulation of cells with an elevated chemoresistance and an increased capacity for self-renewal and metastatic spread. Specifically targeting these cells by adoptive immunotherapy represents a promising strategy to reduce the risk for recurrent disease. This study selected the widely accepted CSC marker CD133 as a target for a chimeric antigen receptor (CAR)-based immunotherapeutic approach to treat ovarian cancer. A lentiviral vector was generated encoding a third-generation anti-CD133-CAR, and clinically used NK92 cells were transduced. These engineered natural killer (NK) cells showed specific killing against CD133-positive ovarian cancer cell lines and primary ovarian cancer cells cultured from sequential ascites harvests. Additionally, specific activation of these engineered NK cells was demonstrated via interferon-gamma secretion assays. To improve clinical efficacy of ovarian cancer treatment, the effect of the chemotherapeutic agent cisplatin was evaluated together with CAR-transduced NK cell treatment. It was demonstrated that NK cells remain cytotoxic and active under cisplatin treatment and, importantly, that sequential treatment with cisplatin followed by CAR-NK cells led to the strongest killing effect. The specific eradication of ovarian CSCs by anti-CD133-CAR expressing NK92 cells represents a promising strategy and, when confirmed in vivo, shall be the basis of future clinical studies with the aim to prevent recurrent disease.


Subject(s)
Antineoplastic Agents/pharmacology , Cytotoxicity, Immunologic , Immunotherapy , Neoplastic Stem Cells/immunology , Ovarian Neoplasms/immunology , Receptors, Antigen/genetics , Recombinant Fusion Proteins , AC133 Antigen/antagonists & inhibitors , AC133 Antigen/immunology , Animals , Antibody Affinity/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression , Gene Order , Genetic Vectors/genetics , Humans , Immunotherapy/methods , Lentivirus/genetics , Mice , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Protein Binding/immunology , Receptors, Antigen/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Transduction, Genetic , Transgenes , Xenograft Model Antitumor Assays
16.
Cancer Res Treat ; 49(4): 1140-1152, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28231426

ABSTRACT

PURPOSE: The selective elimination of cancer stem cells (CSCs) in tumor patients is a crucial goal because CSCs cause drug refractory relapse. To improve the current conventional bispecific immune-engager platform, a 16133 bispecific natural killer (NK) cell engager (BiKE), consisting of scFvs binding FcγRIII (CD16) on NK cells and CD133 on carcinoma cells, was first synthesized and a modified interleukin (IL)-15 crosslinker capable of stimulating NK effector cells was introduced. MATERIALS AND METHODS: DNA shuffling and ligation techniques were used to assemble and synthesize the 1615133 trispecific NK cell engager (TriKE). The construct was tested for its specificity using flow cytometry, cytotoxic determinations using chromium release assays, and lytic degranulation. IL-15-mediated expansion was measured using flow-based proliferation assays. The level of interferon (IFN)-γ release was measured because of its importance in the anti-cancer response. RESULTS: 1615133 TriKE induced NK cell-mediated cytotoxicity and NK expansion far greater than that achieved with BiKE devoid of IL-15. The drug binding and induction of cytotoxic degranulation was CD133+ specific and the anti-cancer activity was improved by integrating the IL-15 cross linker. The NK cell-related cytokine release measured by IFN-γ detection was higher than that of BiKE. NK cytokine release studies showed that although the IFN-γ levels were elevated, they did not approach the levels achieved with IL-12/IL-18, indicating that release was not at the supraphysiologic level. CONCLUSION: 1615133 TriKE enhances the NK cell anti-cancer activity and provides a self-sustaining mechanism via IL-15 signaling. By improving the NK cell performance, the new TriKE represents a highly active drug against drug refractory relapse mediated by CSCs.


Subject(s)
AC133 Antigen/antagonists & inhibitors , Antibody-Dependent Cell Cytotoxicity/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology , T-Cell Antigen Receptor Specificity/immunology , Cell Culture Techniques , Cell Degranulation/immunology , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Genetic Engineering , Humans , Immunotherapy , Interferon-gamma/biosynthesis , Interleukin-15/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism
17.
Mol Cancer ; 16(1): 21, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28137267

ABSTRACT

BACKGROUND: Pilocytic astrocytomas (PAs) are the most common pediatric central nervous system neoplasms. In the majority of cases these tumors are benign and receive favorable prognosis following gross total surgical resection. In patients with progressive or symptomatic tumors, aggressive surgical resection is generally not feasible, thus radiation or chemotherapy are accepted initial or adjuvant interventions. Due to serious long-lasting side-effects, radiation is limited in young children; therefore, chemotherapy is widely practiced as an adjuvant treatment for these patients. However, chemotherapy can promote the emergence of multidrug resistant tumor cells that are more malignant than those of the original tumor. CD133, a putative stem cell marker in normal tissue and malignant brain tumors, enhances multidrug resistant gene 1 (MDR1) expression following chemotherapy in adult malignant glioblastomas. This study examines the relationship between CD133 and MDR1 in pediatric PAs exposed to chemotherapy, with the goal of identifying therapeutic targets that manifest as a result of chemotherapy. METHODS: Slides were obtained for 15 recurrent PAs, seven of which had received chemotherapy prior to surgical treatment for the recurrent tumor. These samples, as well as primary tumor tissue slides from the same patients were used to investigate CD133 and MDR1 expression via immunofluorescence. Archived frozen tissue samples from the same patients were used to examine CD133, MDR1 and PI3K-Akt-NF-κB signaling mediators, via western blot. Two drug resistant pediatric PA cell lines Res186 and Res199 were also used to evaluate the role of CD133 on cell response to cytotoxic therapy. RESULTS: CD133 and MDR1 were co-expressed and their expression was elevated in recurrent PAs from patients that had received chemotherapy, compared to patients that had not received chemotherapy. PI3K-Akt-NF-κB signaling mediator expression was also elevated in recurrent, chemotherapy-treated PA. Suppressing CD133 expression with siCD133 decreased levels of PI3K-Akt-NF-κB signaling mediators and MDR1, while increasing cell chemosensitivity, as indicated by quantification of apoptotic cells following chemotherapy. CONCLUSIONS: CD133 contributes to multidrug resistance by regulating MDR1 levels via the PI3K-Akt-NF-κB signal pathway not only in adult glioblastomas, but also in pediatric PAs. Targeting CD133, adjuvant to conventional chemotherapy may improve outcomes for children with recurrent PA.


Subject(s)
AC133 Antigen/metabolism , Astrocytoma/metabolism , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/metabolism , Up-Regulation , AC133 Antigen/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adolescent , Astrocytoma/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemotherapy, Adjuvant , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Male , NF-kappa B/genetics , Neoadjuvant Therapy , Neoplasm Recurrence, Local/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
18.
Oncotarget ; 7(23): 34430-41, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27144437

ABSTRACT

Gastric cancer remains a disease with a high mortality rate despite of multiple therapeutic strategies. So far, it is very important to develop new treatment approaches to improve current therapeutic efficacy in gastric cancer. Apurinic/apyrimidinic endonuclease (APE1) involves in DNA base excision repair (BER) during DNA damage pathway. APE1 was found to be associated with poor overall survival with gastric cancer patients. In the in vitro experiment, we tested APE1 inhibitor-AT101 could potently inhibit gastric cancer cell growth and further induce cancer cell apoptosis and autophagy through p53-dependent pathway. Downregulation of APE1 by AT101 has ability to suppress gastric cancer cell migration and renewal through inhibition of CD133, Nanog and LC3expression. Based on findings that Her-2 positive expression cases has poor prognosis from our dataset and TCGA database, we investigated the role of AT101 in synergetic efficacy with 5-FU treatment in Her-2 overexpression gastric cancer in vivo, indicating that AT101 is able to enhance 5-FU in the shrinkage of xenograft mice tumor and induction of cell apoptosis. In summary, the data obtained from our study showed APE1 is guided as a potential therapeutic target for gastric cancer. AT101 could be regarded as a potent inhibitor to promote chemotherapeutic sensitivity in patients with gastric cancer.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Fluorouracil/pharmacology , Gossypol/analogs & derivatives , Stomach Neoplasms/drug therapy , AC133 Antigen/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Drug Synergism , Female , Gossypol/pharmacology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/antagonists & inhibitors , Middle Aged , Nanog Homeobox Protein/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
19.
Oncotarget ; 7(28): 43095-43108, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27191269

ABSTRACT

The uptake of (10)boron by tumor cells plays an important role for cell damage in boron neutron capture therapy (BNCT). CD133 is frequently expressed in the membrane of glioma stem cells (GSCs), resistant to radiotherapy and chemotherapy, and represents a potential therapeutic target. To increase (10)boron uptake in GSCs, we created a polyamido amine dendrimer, conjugated CD133 monoclonal antibodies, encapsulating mercaptoundecahydrododecaborate (BSH) in void spaces, and monitored the uptake of the bioconjugate nanoparticles by GSCs in vitro and in vivo. Fluorescence microscopy showed the specific uptake of the bioconjugate nanoparticles by CD133-positive GSCs. Treatment with the biconjugate nanoparticles resulted in a significant lethal effect after neutron radiation due to efficient and CD133-independent cellular targeting and uptake in CD133-expressing GSCs. A significantly longer survival occurred in combination with the biconjugate nanoparticles and BSH compared with BSH alone in human intracranial GBM models employing CD133-positive GSCs xenografts. Our data demonstrated that this bioconjugate nanoparticle targets human CD133-positive GSCs and is a potential boron agent in BNCT.


Subject(s)
AC133 Antigen/antagonists & inhibitors , Antineoplastic Agents, Immunological/therapeutic use , Boron Neutron Capture Therapy/methods , Brain Neoplasms/radiotherapy , Cell Membrane/radiation effects , Glioma/radiotherapy , Neoplastic Stem Cells/radiation effects , AC133 Antigen/metabolism , Animals , Antineoplastic Agents, Immunological/administration & dosage , Borohydrides/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Dendrimers/chemistry , Glioma/drug therapy , Glioma/mortality , Glioma/pathology , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Nanoconjugates/chemistry , Neoplastic Stem Cells/metabolism , Sulfhydryl Compounds/administration & dosage , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...