Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Virus Res ; 345: 199391, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754785

ABSTRACT

Coronaviruses (CoVs) are enveloped single-stranded RNA viruses that predominantly attack the human respiratory system. In recent decades, several deadly human CoVs, including SARS-CoV, SARS-CoV-2, and MERS-CoV, have brought great impact on public health and economics. However, their high infectivity and the demand for high biosafety level facilities restrict the pathogenesis research of CoV infection. Exacerbated inflammatory cell infiltration is associated with poor prognosis in CoV-associated diseases. In this study, we used human CoV 229E (HCoV-229E), a CoV associated with relatively fewer biohazards, to investigate the pathogenesis of CoV infection and the regulation of neutrophil functions by CoV-infected lung cells. Induced pluripotent stem cell (iPSC)-derived alveolar epithelial type II cells (iAECIIs) exhibiting specific biomarkers and phenotypes were employed as an experimental model for CoV infection. After infection, the detection of dsRNA, S, and N proteins validated the infection of iAECIIs with HCoV-229E. The culture medium conditioned by the infected iAECIIs promoted the migration of neutrophils as well as their adhesion to the infected iAECIIs. Cytokine array revealed the elevated secretion of cytokines associated with chemotaxis and adhesion into the conditioned media from the infected iAECIIs. The importance of IL-8 secretion and ICAM-1 expression for neutrophil migration and adhesion, respectively, was demonstrated by using neutralizing antibodies. Moreover, next-generation sequencing analysis of the transcriptome revealed the upregulation of genes associated with cytokine signaling. To summarize, we established an in vitro model of CoV infection that can be applied for the study of the immune system perturbations during severe coronaviral disease.


Subject(s)
Alveolar Epithelial Cells , Induced Pluripotent Stem Cells , Neutrophils , Humans , Neutrophils/immunology , Neutrophils/virology , Induced Pluripotent Stem Cells/virology , Alveolar Epithelial Cells/virology , COVID-19/virology , COVID-19/immunology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , SARS-CoV-2/immunology , Interleukin-8/genetics , Interleukin-8/metabolism
2.
J Chin Med Assoc ; 87(3): 261-266, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38305450

ABSTRACT

BACKGROUND: Leber hereditary optic neuropathy (LHON) is mainly the degeneration of retinal ganglion cells (RGCs) associated with high apoptosis and reactive oxygen species (ROS) levels, which is accepted to be caused by the mutations in the subunits of complex I of the mitochondrial electron transport chain. The treatment is still infant while efforts of correcting genes or using antioxidants do not bring good and consistent results. Unaffected carrier carries LHON mutation but shows normal phenotype, suggesting that the disease's pathogenesis is complex, in which secondary factors exist and cooperate with the primary complex I dysfunction. METHODS: Using LHON patient-specific induced pluripotent stem cells (iPSCs) as the in vitro disease model, we previously demonstrated that circRNA_0087207 had the most significantly higher expression level in the LHON patient-iPSC-derived RGCs compared with the unaffected carrier-iPSC-derived RGCs. To elaborate the underlying pathologies regulated by circRNA_008720 mechanistically, bioinformatics analysis was conducted and elucidated that circRNA_0087207 could act as a sponge of miR-548c-3p and modulate PLSCR1/TGFB2 levels in ND4 mutation-carrying LHON patient-iPSC-derived RGCs. RESULTS: Using LHON iPSC-derived RGCs as the disease-based platform, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis on targeted mRNA of miR-548c-3p showed the connection with apoptosis, suggesting downregulation of miR548c-3p contributes to the apoptosis of LHON patient RGCs. CONCLUSION: We showed that the downregulation of miR548c-3p plays a critical role in modulating cellular dysfunction and the apoptotic program of RGCs in LHON.


Subject(s)
MicroRNAs , Optic Atrophy, Hereditary, Leber , Humans , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/pathology , RNA, Circular/genetics , Mitochondria , Apoptosis , Mutation , MicroRNAs/genetics , MicroRNAs/metabolism , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism
3.
J Chin Med Assoc ; 87(2): 163-170, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38132887

ABSTRACT

BACKGROUND: The potential of induced pluripotent stem cells (iPSCs) in revolutionizing regenerative medicine cannot be overstated. iPSCs offer a profound opportunity for therapies involving cell replacement, disease modeling, and cell transplantation. However, the widespread application of iPSC cellular therapy faces hurdles, including the imperative to regulate iPSC differentiation rigorously and the inherent genetic disparities among individuals. To address these challenges, the concept of iPSC super donors emerges, holding exceptional genetic attributes and advantageous traits. These super donors serve as a wellspring of standardized, high-quality cell sources, mitigating inter-individual variations and augmenting the efficacy of therapy. METHODS: In pursuit of this goal, our study embarked on the establishment of iPSC cell lines specifically sourced from donors possessing the HLA type (A33:03-B58:01-DRB1*03:01). The reprogramming process was meticulously executed, resulting in the successful generation of iPSC lines from these carefully selected donors. Subsequently, an extensive characterization was conducted to comprehensively understand the features and attributes of these iPSC lines. RESULTS: The outcomes of our research were highly promising. The reprogramming efforts culminated in the generation of iPSC lines from donors with the specified HLA type. These iPSC lines displayed a range of distinctive characteristics that were thoroughly examined and documented. This successful generation of iPSC lines from super donors possessing advantageous genetic traits represents a significant stride towards the realization of their potential in therapeutic applications. CONCLUSION: In summary, our study marks a crucial milestone in the realm of regenerative medicine. The establishment of iPSC lines from super donors with specific HLA types signifies a paradigm shift in addressing challenges related to iPSC cellular therapy. The standardized and high-quality cell sources derived from these super donors hold immense potential for various therapeutic applications. As we move forward, these findings provide a solid foundation for further research and development, ultimately propelling the field of regenerative medicine toward new horizons of efficacy and accessibility.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Cellular Reprogramming , Cell Differentiation , Cell- and Tissue-Based Therapy
4.
Biochem Pharmacol ; 206: 115289, 2022 12.
Article in English | MEDLINE | ID: mdl-36241092

ABSTRACT

Colorectal cancer (CRC) is a leading cause and mortality worldwide. Aurora A and haspin kinases act pivotal roles in mitotic progression. However, the blockage of Aurora A and Haspin for CRC therapy is still unclear. Here we show that the Haspin and p-H3T3 protein levels were highly expressed in CRC tumor tissues of clinical patients. Overexpression of Haspin increased the protein levels of p-H3T3 and survivin in human CRC cells; conversely, the protein levels of p-H3T3 and survivin were decreased by the Haspin gene knockdown. Moreover, the gene knockdown of Aurora A induced abnormal chromosome segregation, mitotic catastrophe, and cell growth inhibition. Combined targeted by co-treatment of CHR6494, a Haspin inhibitor, and MLN8237, an Aurora A inhibitor, enhanced apoptosis and CRC tumor inhibition. MLN8237 and CHR6494 induced abnormal chromosome segregation and mitotic catastrophe. Meanwhile, MLN8237 and CHR6494 inhibited survivin protein levels but conversely induced p53 protein expression. Ectopic survivin expression by transfection with a survivin-expressed vector resisted the cell death in the MLN8237- and CHR6494-treated cells. In contrast, the existence of functional p53 increased the apoptotic levels by treatment with MLN8237 and CHR6494. Co-treatment of CHR6494 and MLN8237 enhanced the blockage of human CRC xenograft tumors in nude mice. Taken together, co-inhibition of Aurora A and Haspin enhances survivin inhibition, p53 pathway induction, mitotic catastrophe, apoptosis and tumor inhibition that may provide a potential strategy for CRC therapy.


Subject(s)
Aurora Kinase A , Colorectal Neoplasms , Survivin , Tumor Suppressor Protein p53 , Animals , Humans , Mice , Apoptosis , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Mice, Nude , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Survivin/genetics , Survivin/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics
5.
Biomedicines ; 10(4)2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35453537

ABSTRACT

Backgrounds: Leber's hereditary optic neuropathy (LHON) is known as an inherited retinal disorder characterized by the bilateral central vision loss and degeneration of retinal ganglion cells (RGCs). Unaffected LHON carriers are generally asymptomatic, suggesting that certain factors may contribute to the disease manifestations between carriers and patients who carry the same mutated genotypes. Methods: We first aimed to establish the iPSC-differentiated RGCs from the normal healthy subject, the carrier, and the LHON patient and then compared the differential expression profile of circular RNAs (CircRNAs) among RGCs from these donors in vitro. We further overexpressed or knocked down the most upregulated circRNA to examine whether this circRNA contributes to the distinct phenotypic manifestations between the carrier- and patient-derived RGCs. Results: iPSCs were generated from the peripheral blood cells from the healthy subject, the carrier, and the LHON patient and successfully differentiated into RGCs. These RGCs carried equivalent intracellular reactive oxygen species, but only LHON-patient iPSC-derived RGCs exhibited remarkable apoptosis. Next-generation sequencing and quantitative real-time PCR revealed the circRNA hsa_circ_0087207 as the most upregulated circRNA in LHON-patient iPSC-derived RGCs. Overexpression of hsa_circ_0087207 increased the apoptosis in carrier iPSC-derived RGCs, while knockdown of hsa_circ_0087207 attenuated the apoptosis in LHON-patient iPSC-derived RGCs. Predicted by bioinformatics approaches, hsa_circ_0087207 acts as the sponge of miR-665 to induce the expression of a variety of apoptosis-related genes in LHON patient iPSC-derived RGCs. Conclusions: Our data indicated that hsa_circ_0087207 upregulation distinguishes the disease phenotype manifestations between iPSC-derived RGCs generated from the LHON patient and carrier. Targeting the hsa_circ_0087207/miR-665 axis might hold therapeutic promises for the treatment of LHON.

6.
Cells ; 8(6)2019 06 21.
Article in English | MEDLINE | ID: mdl-31234430

ABSTRACT

The mitochondrial genetic disorder, Leber's hereditary optic neuropathy (LHON), is caused by a mutation in MT-ND4 gene, encoding NADH dehydrogenase subunit 4. It leads to the progressive death of retinal ganglion cells (RGCs) and causes visual impairment or even blindness. However, the precise mechanisms of LHON disease penetrance and progression are not completely elucidated. Human-induced pluripotent stem cells (hiPSCs) offer unique opportunities to investigate disease-relevant phenotypes and regulatory mechanisms underlying LHON pathogenesis at the cellular level. In this study, we successfully generated RGCs by differentiation of LHON patient-specific hiPSCs. We modified the protocol of differentiation to obtain a more enriched population of single-cell RGCs for LHON study. Based on assessing morphology, expression of specific markers and electrophysiological activity, we found that LHON-specific hiPSC-derived were more defective in comparison with normal wild-type RGCs. Based on our previous study, whereby by using microarray analysis we identified that the components of glutamatergic synapse signaling pathway were significantly downregulated in LHON-specific RGCs, we focused our study on glutamate-associated α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors. We found that the protein expression levels of the subunits of the AMPA receptor, GluR1 and GluR2, and their associated scaffold proteins were decreased in LHON-RGCs. By performing the co-immunoprecipitation assay, we found several differences in the efficiencies of interaction between AMPA subunits and scaffold proteins between normal and LHON-specific RGCs.


Subject(s)
Glutamic Acid/pharmacology , Induced Pluripotent Stem Cells/metabolism , Optic Atrophy, Hereditary, Leber/metabolism , Optic Atrophy, Hereditary, Leber/pathology , Receptors, AMPA/metabolism , Retinal Ganglion Cells/metabolism , Signal Transduction , Base Sequence , Cell Differentiation/drug effects , Down-Regulation/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Protein Binding/drug effects , Retinal Ganglion Cells/drug effects , Signal Transduction/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
7.
Sci Rep ; 7(1): 2172, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28526879

ABSTRACT

Colorectal cancers (CRCs) are a critical health issue worldwide. Cancer stem cell (CSC) lineages are associated with tumour transformation, progression, and malignant transformation. However, how lineages are transformed and how chemoresistance is acquired by CRCs remain largely unknown. In this report, we demonstrated that the RNA-binding protein Musashi-1 enhanced the development of CD44+ colorectal CSCs and triggered the formation of anti-apoptotic stress granules (SGs). Our results indicated that CD44+ CSC lineage-specific induction of tumour malignancies was controlled by Musashi-1. In addition, Musashi-1 formed SGs when CRC cell lines were treated with 5-fluorouracil. The C-terminal domain of Musashi-1 was critical for recruitment of Musashi-1 into SGs. Intracellular Musashi-1 SGs enhanced the chemoresistance of CRCs. Analysis of clinical CRC samples indicated that Musashi-1 expression was prominent in CRC stage IIA and IIB. In summary, we demonstrated that Musashi-1, a stemness gene, is a critical modulator that promotes the development of CD44+ colorectal CSCs and also enhances CRC chemoresistance via formation of SGs. Our findings elucidated a novel mechanism of CRC chemoresistance through increased anti-apoptotic effects via Musashi-1-associated SGs.


Subject(s)
Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Biomarkers , Cell Line, Tumor , Cell Lineage , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Fluorescent Antibody Technique , Fluorouracil/pharmacology , Humans , Immunohistochemistry , Neoplasm Staging , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics
8.
Eur J Pharm Biopharm ; 109: 165-173, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27793756

ABSTRACT

Lung cancer kills more humans than any other cancer and multidrug resistance (MDR) in cancer stem-like cells (CSC) is emerging as a reason for failed treatments. One concept that addresses this root cause of treatment failure is the utilization of nanoparticles to simultaneously deliver dual drugs to cancer cells with synergistic performance, easy to envision - hard to achieve. (1) It is challenging to simultaneously load drugs of highly different physicochemical properties into one nanoparticle, (2) release kinetics may differ between drugs and (3) general requirements for biomedical nanoparticles apply. Here self-assembled nanoparticles of amphiphilic carboxymethyl-hexanoyl chitosan (CHC) were shown to present nano-microenvironments enabling simultaneous loading of hydrophilic and hydrophobic drugs. This was expanded into a dual-drug nano-delivery system to treat lung CSC. CHC nanoparticles were loaded/chemically modified with the anticancer drug cisplatin and the MDR-suppressing Chinese herbal extract demethoxycurcumin, followed by biofunctionalization with CD133 antibody for enhanced uptake by lung CSC, all in a feasible one-pot preparation. The nanoparticles were characterized with regard to chemistry, size, zeta potential and drug loading/release. Biofunctionalized and non-functionalized nanoparticles were investigated for uptake by lung CSC. Subsequently the cytotoxicity of single and dual drugs, free in solution or in nanoparticles, was evaluated against lung CSC at different doses. From the dose response at different concentrations the degree of synergy was determined through Chou-Talalay's Plot. The biofunctionalized nanoparticles promoted synergistic effects between the drugs and were highly effective against MDR lung CSC. The efficacy and feasible one-pot preparation suggests preclinical studies using relevant disease models to be justified.


Subject(s)
Chitosan/chemistry , Cisplatin/administration & dosage , Curcumin/analogs & derivatives , Drug Resistance, Neoplasm , Lung Neoplasms/metabolism , Nanoparticles/chemistry , A549 Cells , AC133 Antigen/metabolism , Antineoplastic Agents/administration & dosage , Biomarkers/metabolism , Chitosan/analogs & derivatives , Colloids/chemistry , Curcumin/administration & dosage , Diarylheptanoids , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Delivery Systems , Drug Liberation , Drug Resistance, Multiple , Drugs, Chinese Herbal/administration & dosage , Humans , Kinetics , Neoplastic Stem Cells/drug effects , Photoelectron Spectroscopy , Spectroscopy, Fourier Transform Infrared
9.
Oncotarget ; 6(3): 1750-68, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25638155

ABSTRACT

Atypical teratoid/rhabdoid tumor (ATRT) is a malignant pediatric brain tumor with great recurrence after complete surgery and chemotherapy. Here, we demonstrate that cisplatin treatment selects not only for resistance but also for a more oncogenic phenotype characterized by high self-renewal and invasive capabilities. These phenomena are likely due to STAT3 upregulatoin which occurred simultaneously with higher expression of Snail, an activator of epithelial-mesenchymal transition (EMT), in ATRT-CisR cells. STAT3 knockdown effectively suppressed Snail expression and blocked motility and invasion in ATRT-CisR cells, while overexpressing Snail reversed these effects. Chromatin immunoprecipitation assay indicated that STAT3 directly bound to Snail promoter. Moreover, STAT3 knockdown effectively suppressed cancer stem-like properties, synergistically enhanced the chemotherapeutic effect, and significantly improved survival rate in ATRT-CisR-transplanted immunocompromised mice. Finally, immunohistochemistrical analysis showed that STAT3 and Snail were coexpressed at high levels in recurrent ATRT tissues. Thus, the STAT3/Snail pathway plays an important role in oncogenic resistance, rendering cells not only drug-resistant but also increasingly oncogenic (invasion, EMT and recurrence). Therefore, the STAT3/Snail could be a target for ATRT treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Cell Movement/physiology , Cisplatin/pharmacology , Rhabdoid Tumor/pathology , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Heterografts , Humans , Male , Mice , Mice, SCID , Rhabdoid Tumor/drug therapy , Rhabdoid Tumor/metabolism , Signal Transduction , Snail Family Transcription Factors
10.
Mol Cell ; 52(5): 693-706, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24332177

ABSTRACT

Epigenetic regulation plays a critical role in glioblastoma (GBM) tumorigenesis. However, how microRNAs (miRNAs) and cytokines cooperate to regulate GBM tumor progression is still unclear. Here, we show that interleukin-6 (IL-6) inhibits miR142-3p expression and promotes GBM propagation by inducing DNA methyltransferase 1-mediated hypermethylation of the miR142-3p promoter. Interestingly, miR142-3p also suppresses IL-6 secretion by targeting the 3' UTR of IL-6. In addition, miR142-3p also targets the 3' UTR and suppresses the expression of high-mobility group AT-hook 2 (HMGA2), leading to inhibition of Sox2-related stemness. We further show that HMGA2 enhances Sox2 expression by directly binding to the Sox2 promoter. Clinically, GBM patients whose tumors present upregulated IL-6, HMGA2, and Sox2 protein expressions and hypermethylated miR142-3p promoter also demonstrate poor survival outcome. Orthotopic delivery of miR142-3p blocks IL-6/HMGA2/Sox2 expression and suppresses stem-like properties in GBM-xenotransplanted mice. Collectively, we discovered an IL-6/miR142-3p feedback-loop-dependent regulation of GBM malignancy that could be a potential therapeutic target.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Interleukin-6/genetics , MicroRNAs/genetics , 3' Untranslated Regions , Animals , Base Sequence , Cell Line, Tumor , DNA Methylation , Epigenesis, Genetic , Female , HMGA2 Protein/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Molecular Sequence Data , Promoter Regions, Genetic , SOXB1 Transcription Factors/genetics , Up-Regulation
11.
Transplantation ; 96(11): 958-65, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23989473

ABSTRACT

BACKGROUND: Allergic disorders have increased substantially in recent years. Asthma is characterized by airway damage and remodeling. Reprogramming induced pluripotent stem cells (iPSCs) from adult somatic cells transfected by Oct-4/Sox-2/Klf-4, but not c-Myc, has shown the potential of embryonic-like cells. These cells have potential for multilineage differentiation and provide a resource for stem cell-based utility. However, the therapeutic potential of iPSCs without c-Myc (iPSC-w/o-c-Myc) in allergic diseases and airway hyperresponsiveness has not been investigated. The aim of this study was to evaluate the therapeutic effect of iPSC-w/o-c-Myc transplantation in a murine asthma model. METHODS: BALB/c mice were sensitized with alum-adsorbed ovalbumin (OVA) and then challenged with aerosolized OVA. Phosphate-buffered saline or iPSC-w/o-c-Myc was then intravenously injected after inhalation. Serum allergen-specific antibody levels, airway hyperresponsiveness, cytokine levels in spleen cells and bronchoalveolar lavage fluid (BALF), and cellular distribution in BALF were then examined. RESULTS: Treatment with iPSC-w/o-c-Myc effectively suppressed both Th1 and Th2 antibody responses, which was characterized by reduction in serum allergen-specific IgE, IgG, IgG1, and IgG2a levels as well as in interleukin-5 and interferon-γ levels in BALF and in OVA-incubated splenocytes. Meanwhile, regulatory cytokine, interleukin-10, was enhanced. Transplantation of iPSC-w/o-c-Myc also significantly attenuated cellular infiltration in BALF and allergic airway hyperresponsiveness. However, no tumor formation was observed 6 months after transplantation. CONCLUSIONS: Administration of iPSC-w/o-c-Myc not only inhibited Th1 inflammatory responses but also had therapeutic effects on systemic allergic responses and airway hyperresponsiveness. iPSC-w/o-c-Myc transplantation may be a potential modality for treating allergic reactions and bronchial asthma.


Subject(s)
Asthma/prevention & control , Bronchial Hyperreactivity/prevention & control , Induced Pluripotent Stem Cells/transplantation , Lung/immunology , Proto-Oncogene Proteins c-myc/deficiency , Th2 Cells/immunology , Animals , Asthma/immunology , Asthma/physiopathology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Immunoglobulin E/metabolism , Induced Pluripotent Stem Cells/metabolism , Inflammation Mediators/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lung/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Ovalbumin , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Spleen/immunology , Th1 Cells/immunology , Time Factors , Transfection
12.
Cancer Res ; 73(11): 3425-40, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23548270

ABSTRACT

ALDH1(+)CD44(+) cells are putative tumor-initiating cells (TIC) in head and neck squamous cell carcinomas (HNC). miR-145 regulates tumorigenicity in various cancers but the breadth of its mechanistic contributions and potential therapeutic applications are not completely known. Here, we report that ALDH1(+)CD44(+)-HNC cells express reduced levels of miR145. SPONGE-mediated inhibition of miR-145 (Spg-miR145) was sufficient to drive tumor-initiating characteristics in non-TICs/ALDH1(-)CD44-negative HNC cells. Mechanistic analyses identified SOX9 and ADAM17 as two novel miR145 targets relevant to this process. miR-145 expression repressed TICs in HNC in a manner associated with SOX9 interaction with the ADAM17 promoter, thereby activating ADAM17 expression. Notably, the SOX9/ADAM17 axis dominated the TIC-inducing activity of miR-145. Either miR-145 suppression or ADAM17 overexpression in non-TICs/ALDH1(-)CD44(-)-HNC cells increased expression and secretion of interleukin (IL)-6 and soluble-IL-6 receptor (sIL-6R). Conversely, conditioned medium from Spg-miR145-transfected non-TICs/ALDH1(-)CD44(-)-HNC cells was sufficient to confer tumor-initiating properties in non-TICs/ALDH1(-)CD44(-)-HNC and this effect could be abrogated by an IL-6-neutralizing antibody. We found that curcumin administration increased miR-145 promoter activity, thereby decreasing SOX9/ADAM17 expression and eliminating TICs in HNC cell populations. Delivery of lentivral-miR145 or orally administered curcumin blocked tumor progression in HNC-TICs in murine xenotransplant assays. Finally, immunohistochemical analyses of patient specimens confirmed that an miR-145(low)/SOX9(high)/ADAM17(high) phenotype correlated with poor survival. Collectively, our results show how miR-145 targets the SOX9/ADAM17 axis to regulate TIC properties in HNC, and how altering this pathway may partly explain the anticancer effects of curcumin. By inhibiting IL-6 and sIL-6R as downstream effector cytokines in this pathway, miR-145 seems to suppress a paracrine signaling pathway in the tumor microenvironment that is vital to maintain TICs in HNC.


Subject(s)
ADAM Proteins/metabolism , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Interleukin-6/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , SOX9 Transcription Factor/metabolism , 3' Untranslated Regions , ADAM Proteins/genetics , ADAM17 Protein , Animals , Base Sequence , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Growth Processes/genetics , Cell Line, Tumor , Curcumin/pharmacology , Epithelial-Mesenchymal Transition , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Heterografts , Humans , Interleukin-6/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Receptors, Interleukin-6/metabolism , SOX9 Transcription Factor/genetics , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
13.
Shock ; 39(3): 261-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23364435

ABSTRACT

Chronic lung diseases cause serious morbidity and mortality, and effective treatments are limited. Induced pluripotent stem cells (iPSCs) lacking the reprogramming factor c-Myc (3-gene iPSCs) can be used as ideal tools for cell-based therapy because of their low level of tumorigenicity. In this study, we investigated whether 3-gene iPSC transplantation could rescue bleomycin-induced pulmonary fibrosis. After the induction of pulmonary inflammation and fibrosis via intratracheal delivery of bleomycin sulfate, mice were i.v. injected with 3-gene iPSCs or conditioned medium (iPSC-CM) at 24 h after bleomycin treatment. Administration of either 3-gene iPSCs or iPSC-CM significantly attenuated collagen content and myeloperoxidase activity, diminished neutrophil accumulation, and rescued pulmonary function and recipient survival after bleomycin treatment. Notably, both treatments reduced the levels of inflammatory cytokines and chemokines, including interleukin 1 (IL-1), IL-2, IL-10, tumor necrosis factor-α, and monocyte chemotactic protein 1 yet increased the production of the antifibrotic chemokine interferon-γ-induced protein 10 (IP-10) in bleomycin-injured lungs. Furthermore, IP-10 neutralization via treatment with IP-10-neutralizing antibodies ameliorated the reparative effect of either 3-gene iPSCs or iPSC-CM on collagen content, neutrophil and monocyte accumulation, pulmonary fibrosis, and recipient survival. Intravenous delivery of 3-gene iPSCs/iPSC-CM alleviated the severity of histopathologic and physiologic impairment in bleomycin-induced lung fibrosis. The protective mechanism was partially mediated by the early moderation of inflammation, reduced levels of cytokines and chemokines that mediate inflammation and fibrosis, and an increased production of antifibrotic IP-10 in the injured lungs.


Subject(s)
Chemokine CXCL10/biosynthesis , Induced Pluripotent Stem Cells/transplantation , Pneumonia/therapy , Pulmonary Fibrosis/therapy , Animals , Bleomycin , Culture Media, Conditioned , Cytokines/biosynthesis , Disease Models, Animal , Genes, myc , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Peroxidase/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
14.
J Exp Med ; 210(1): 85-98, 2013 Jan 14.
Article in English | MEDLINE | ID: mdl-23277454

ABSTRACT

Poly(ADP-ribose) polymerase 1 (Parp1) catalyzes poly(ADP-ribosylation) (PARylation) and induces replication networks involved in multiple nuclear events. Using mass spectrometry and Western blotting, Parp1 and PARylation activity were intensively detected in induced pluripotent stem cells (iPSCs) and embryonic stem cells, but they were lower in mouse embryonic fibroblasts (MEFs) and differentiated cells. We show that knockdown of Parp1 and pharmacological inhibition of PARylation both reduced the efficiency of iPSC generation induced by Oct4/Sox2/Klf4/c-Myc. Furthermore, Parp1 is able to replace Klf4 or c-Myc to enhance the efficiency of iPSC generation. In addition, mouse iPSCs generated from Oct4/Sox2/Parp1-overexpressing MEFs formed chimeric offspring. Notably, the endogenous Parp1 and PARylation activity was enhanced by overexpression of c-Myc and repressed by c-Myc knockdown. A chromatin immunoprecipitation assay revealed a direct interaction of c-Myc with the Parp1 promoter. PAR-resin pulldown, followed by proteomic analysis, demonstrated high levels of PARylated Chd1L, DNA ligase III, SSrp1, Xrcc-6/Ku70, and Parp2 in pluripotent cells, which decreased during the differentiation process. These data show that the activation of Parp1, partly regulated by endogenous c-Myc, effectively promotes iPSC production and helps to maintain a pluripotent state by posttranslationally modulating protein PARylation.


Subject(s)
Cellular Reprogramming , Genes, myc , Induced Pluripotent Stem Cells/physiology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Antigens, Nuclear/metabolism , Cell Differentiation , Cells, Cultured , Chimera , DNA Ligase ATP , DNA Ligases/metabolism , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockdown Techniques , High Mobility Group Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Ku Autoantigen , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly-ADP-Ribose Binding Proteins , Promoter Regions, Genetic , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Xenopus Proteins
15.
Sci Rep ; 2: 584, 2012.
Article in English | MEDLINE | ID: mdl-22900142

ABSTRACT

Mounting evidence links cancers possessing stem-like properties with worse prognosis. Network biology with signal processing mechanics was explored here using expression profiles of a panel of tumor stem-like cells (TSLCs). The profiles were compared to their parental tumor cells (PTCs) and the human embryonic stem cells (hESCs), for the identification of gene chromobox homolog 5, CBX5, as a potential target for lung cancer. CBX5 was found to regulate the stem-like properties of lung TSLCs and was predictive of lung cancer prognosis. The investigation was facilitated by finding target genes based on modeling epistatic signaling mechanics via a predictive and scalable network-based survival model. Topologically-weighted measurements of CBX5 were synchronized with those of BIRC5, DNMT1, E2F1, ESR1, MLH1, MSH2, RB1, SMAD1 and TAF5. We validated our findings in another Taiwanese lung cancer cohort, as well as in knockdown experiments using sh-CBX5 RNAi both in vitro and in vivo.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Animals , Cell Transformation, Neoplastic/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunophenotyping , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Annotation , RNA Interference , Reproducibility of Results , Transcription, Genetic
16.
Curr Gene Ther ; 12(4): 275-84, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22856602

ABSTRACT

RNA interference (RNAi) is a collection of small RNA-directed mechanisms that result in sequence-specific inhibition of gene expression. RNAi delivery has demonstrated promising efficacy in the treatment of genetic disorders in cancer. Although viral vectors are currently the most efficient systems for gene therapy, potent immunogenicity, mutagenesis, and the biohazards of viral vectors remain their major risks. Various non-viral delivery vectors have been developed to provide a safer approach for gene delivery, including polymers, peptides, liposomes, and nanoparticles. However, some concerns and challenges of these non-viral gene delivery approaches remain to be overcome. In this review, we summarize the recent progress in the development of non-viral systems delivering RNAi and the currently available preclinical and clinical data, and discuss the challenges and future directions in cancer therapy.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Neoplasms , RNA Interference , RNA, Small Interfering , Gene Expression Regulation, Neoplastic , Genetic Vectors , Humans , Liposomes , Nanoparticles , Neoplasms/genetics , Neoplasms/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use
17.
Cell Transplant ; 21(12): 2569-85, 2012.
Article in English | MEDLINE | ID: mdl-22507855

ABSTRACT

Induced pluripotent stem (iPS) cells have potential for multilineage differentiation and provide a resource for stem cell-based treatment. However, the therapeutic effect of iPS cells on acute kidney injury (AKI) remains uncertain. Given that the oncogene c-Myc may contribute to tumorigenesis by causing genomic instability, herein we evaluated the therapeutic effect of iPS cells without exogenously introduced c-Myc on ischemia-reperfusion (I/R)-induced AKI. As compared with phosphate-buffered saline (PBS)-treated group, administration of iPS cells via intrarenal arterial route into kidneys improved the renal function and attenuated tubular injury score at 48 h after ischemia particularly at the dose of 5 × 10(5) iPS cells. However, a larger number of iPS cells (5 × 10(7) per rat) diminished the therapeutic effects for AKI and profoundly reduced renal perfusion detected by laser Doppler imaging in the reperfusion phase. In addition, the green fluorescence protein-positive iPS cells mobilized to the peritubular area at 48 h following ischemia, accompanied by a significant reduction in infiltration of macrophages and apoptosis of tubular cells, and a remarkable enhancement in endogenous tubular cell proliferation. Importantly, transplantation of iPS cells reduced the expression of oxidative substances, proinflammatory cytokines, and apoptotic factors in I/R kidney tissues and eventually improved survival in rats of ischemic AKI. Six months after transplantation in I/R rats, engrafted iPS cells did not result in tumor formation in kidney and other organs. In summary, considering the antioxidant, anti-inflammatory, and antiapoptotic properties of iPS cells without c-Myc, transplantation of such cells may be a treatment option for ischemic AKI.


Subject(s)
Acute Kidney Injury/therapy , Induced Pluripotent Stem Cells/transplantation , Inflammation/pathology , Oxidative Stress , Proto-Oncogene Proteins c-myc/metabolism , Acute Kidney Injury/complications , Acute Kidney Injury/mortality , Animals , Apoptosis , Blood Urea Nitrogen , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Down-Regulation , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-myc/genetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Survival Rate
18.
Biomaterials ; 33(14): 3693-709, 2012 May.
Article in English | MEDLINE | ID: mdl-22361100

ABSTRACT

Metastasis is the major cause of high mortality in head and neck squamous cell carcinoma (HNSCC), in which HNSCC-derived cancer stem cells (CSCs) may be involved. Several reports have coupled non-viral gene delivery with RNA interference (RNAi) to target specific genes in cancer cells. However, the delivery efficiency of RNAi is limited and remained to be improved. Moreover, the therapeutic effect of non-viral gene delivery approaches on HNSCC-derived CSCs is still uncertain. In this study, we found that EZH2 and Oct4 are upregulated in HNSCC-derived ALDH1+/CD44+ CSC-like cells. Polyurethane-short branch PEI (PU-PEI)-based administration of double-stranded DNA (dsDNA) encoding small interfering RNA (siRNA) against EZH2 and Oct4 (siEZH2/siOct4) led to partial anti-cancer capacity and mild suppression of CSC-like properties. By pre-conjugation of nuclear localization signal (NLS) to siRNA-expressing dsDNA, the anti-cancer efficacy was enhanced due to elevated nuclear delivery. Notably, the NLS-preconjugated siEZH2/siOct4 constructs remarkably repressed epithelial-mesenchymal transdifferentiation (EMT) and radioresistance in ALDH1+/CD44+ CSC-like cells, in which Wnt5A and CyclinD1 may be involved respectively. We furthermore demonstrated that this improved method was capable of reducing tumor growth and metastasis in vivo. Our findings may provide a feasible non-viral gene delivery method to eradicate HNSCC-derived CSCs and improve HNSCC therapy.


Subject(s)
Carcinoma, Squamous Cell/therapy , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Head and Neck Neoplasms/therapy , Neoplastic Stem Cells/metabolism , Nuclear Localization Signals/genetics , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/genetics , RNA Interference , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Animals , Base Sequence , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Transdifferentiation/genetics , Enhancer of Zeste Homolog 2 Protein , Epithelial-Mesenchymal Transition/genetics , Gene Transfer Techniques , Genetic Vectors , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Male , Materials Testing , Mice , Mice, Nude , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 2 , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
19.
J Control Release ; 159(2): 240-50, 2012 Apr 30.
Article in English | MEDLINE | ID: mdl-22285547

ABSTRACT

The high invasiveness and frequent recurrence of lung adenocarcinoma (LAC) are major reasons for treatment failures and poor prognoses. Alterations in microRNAs (miRNAs) expression have been shown in lung cancers. Recent reports have demonstrated that tumors contain a small subpopulation of cancer stem cells (CSCs) that possesses self-renewing capacity and is responsible for tumor malignancy including metastasis, relapse, and chemoradioresistance. However, a miRNAs-based therapeutic approach in LAC-associated CSCs (LAC-CSCs) is still blurred. Using miRNA/mRNA-microarray and Quantitative RT-PCR, we found that the expression of miR145 is negatively correlated with the levels of Oct4/Sox2/Fascin1 in LAC patient specimens, and an Oct4(high)Sox2(high)Fascin1(high)miR145(low) phenotype predicted poor prognosis. We enriched LAC-CSCs by side population sorting or identification of CD133 markers and found that LAC-CSCs exhibited low miR145 and high Oct4/Sox2/Fascin1 expression, CSC-like properties, and chemoradioresistance. To clarify the role of miR145, we used a polyurethane-short branch-polyethylenimine (PU-PEI) as the vehicle to deliver miR145 into LAC-CSCs. PU-PEI-mediated miR145 delivery reduced CSC-like properties, and improved chemoradioresistance in LAC-CSCs by directly targeting Oct4/Sox2/Fascin1. Importantly, the repressive effect of miR145 on tumor metastasis was mediated by inhibiting the epithelial-mesenchymal transdifferentiation (EMT) and metastastic ability, partially by regulating Oct4/Sox2/Fascin1, Tcf4, and Wnt5a. Finally, in vivo study showed that PU-PEI-mediated miR145 delivery to xenograft tumors reduced tumor growth and metastasis, sensitized tumors to chemoradiotherapies, and prolonged the survival times of tumor-bearing mice. Our results demonstrated that miR145 acts as a switch regulating lung CSC-like and EMT properties, and provide insights into the clinical prospect of miR145-based therapies for malignant lung cancers.


Subject(s)
Adenocarcinoma/drug therapy , Drug Carriers/chemistry , Epithelial-Mesenchymal Transition/drug effects , Gene Transfer Techniques , Lung Neoplasms/drug therapy , MicroRNAs/administration & dosage , Neoplastic Stem Cells/drug effects , Polyethyleneimine/chemistry , Polyurethanes/chemistry , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cations , Cell Culture Techniques , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Survival/drug effects , Cell Survival/genetics , Cell Transdifferentiation , Cells, Cultured , Epithelial-Mesenchymal Transition/genetics , Gamma Rays , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/therapeutic use , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Tumor Stem Cell Assay
20.
Biomaterials ; 33(5): 1462-76, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22098779

ABSTRACT

Glioblastomas (GBMs) are the most common primary brain tumors with poor prognosis. CD133 has been considered a putative marker of cancer stem cells (CSCs) in malignant cancers, including GBMs. MicroRNAs (miRNAs), highly conserved small RNA molecules, may target oncogenes and have potential as a therapeutic strategy against cancer. However, the role of miRNAs in GBM-associated CSCs remains mostly unclear. In this study, our miRNA/mRNA-microarray and RT-PCR analysis showed that the expression of miR145 (a tumor-suppressive miRNA) is inversely correlated with the levels of Oct4 and Sox2 in GBM-CD133(+) cells and malignant glioma specimens. We demonstrated that miR145 negatively regulates GBM tumorigenesis by targeting Oct4 and Sox2 in GBM-CD133(+). Using polyurethane-short branch polyethylenimine (PU-PEI) as a therapeutic-delivery vehicle, PU-PEI-mediated miR145 delivery to GBM-CD133(+) significantly inhibited their tumorigenic and CSC-like abilities and facilitated their differentiation into CD133(-)-non-CSCs. Furthermore, PU-PEI-miR145-treated GBM-CD133(+) effectively suppressed the expression of drug-resistance and anti-apoptotic genes and increased the sensitivity of the cells to radiation and temozolomide. Finally, the in vivo delivery of PU-PEI-miR145 alone significantly suppressed tumorigenesis with stemness, and synergistically improved the survival rate when used in combination with radiotherapy and temozolomide in orthotopic GBM-CD133(+)-transplanted immunocompromised mice. Therefore, PU-PEI-miR145 is a novel therapeutic approach for malignant brain tumors.


Subject(s)
Drug Resistance, Neoplasm , Glioblastoma/genetics , Glioblastoma/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , Polyethyleneimine/analogs & derivatives , Polyurethanes/chemistry , Radiation Tolerance , 3' Untranslated Regions/genetics , Aged , Base Sequence , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Transfer Techniques , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Male , MicroRNAs/genetics , Middle Aged , Molecular Sequence Data , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Polyethyleneimine/chemical synthesis , Polyethyleneimine/chemistry , Polyurethanes/chemical synthesis , Radiation Tolerance/drug effects , SOXB1 Transcription Factors/metabolism , Temozolomide
SELECTION OF CITATIONS
SEARCH DETAIL
...