Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Article in English | MEDLINE | ID: mdl-38522903

ABSTRACT

BACKGROUND: Perfluorooctanoic acid (PFOA) is one of the major per- and polyfluoroalkyl substances. The role of ATP-binding cassette (ABC) transporters in PFOA toxicokinetics is unknown. METHODS: In this study, two ABC transporters, ABCB1 and ABCB4, were examined in mice with single intravenous PFOA administration (3.13 µmol/kg). To identify candidate renal PFOA transporters, we used a microarray approach to evaluate changes in gene expression of various kidney transporters in Abcb4 null mice. RESULTS: Biliary PFOA concentrations were lower in Abcb4 null mice (mean ± standard deviation: 0.25 ± 0.12 µg/mL) than in wild-type mice (0.87 ± 0.02 µg/mL). Immunohistochemically, ABCB4 expression was confirmed at the apical region of hepatocytes. However, renal clearance of PFOA was higher in Abcb4 null mice than in wild-type mice. Among 642 solute carrier and ABC transporters, 5 transporters showed significant differences in expression between wild-type and Abcb4 null mice. These candidates included two major xenobiotic transporters, multidrug resistance 1 (Abcb1) and organic anion transporter 3 (Slc22a8). Abcb1 mRNA levels were higher in Abcb4 null mice than in wild-type mice in kidney. In Abcb4 null mice, Abcb1b expression was enhanced in proximal tubules immunohistochemically, while that of Slc22a8 was not. Finally, in Abcb1a/b null mice, there was a significant decrease in the renal clearance of PFOA (0.69 ± 0.21 vs 1.1 mL ± 0.37/72 h in wild-type mice). A homology search of ABCB1 showed that several amino acids are mutated in humans compared with those in rodents and monkeys. CONCLUSIONS: These findings suggest that, in the mouse, Abcb4 and Abcb1 are excretory transporters of PFOA into bile and urine, respectively.


Subject(s)
Caprylates , Fluorocarbons , Hepatobiliary Elimination , Humans , Mice , Animals , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Fluorocarbons/toxicity , Fluorocarbons/metabolism , Kidney , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism
2.
Diabetes Ther ; 13(11-12): 1847-1860, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36136238

ABSTRACT

INTRODUCTION: The spread of coronavirus disease 2019 (COVID-19) is having a profound effect on global health. In this study, we investigated early predictors of severe prognosis from the perspective of liver injury and risk factors for severe liver injury in patients with COVID-19. METHODS: We examined prognostic markers and risk factors for severe liver injury by analyzing clinical data measured throughout the course of the illness and the disease severity of 273 patients hospitalized for COVID-19. We assessed liver injury on the basis of aminotransferase concentrations and fibrosis-4 (FIB-4) index on admission, peak aminotransferase concentration during hospitalization, aminotransferase peak-to-average ratio, and albumin and total bilirubin concentrations. Furthermore, we analyzed age, aspartate aminotransferase (AST) concentrations, FIB-4 index on admission, hypertension, diabetes mellitus (DM), dyslipidemia, cerebral infarction, myocardial infarction, and body mass index as mortality risk factors. RESULTS: We identified advanced age as a risk factor. Among biochemical variables, AST concentration and FIB-4 index on admission were associated with high mortality. AST on admission and peak AST during hospitalization were significantly higher in the non-surviving (n = 45) than the discharged group (n = 228). Multivariable Cox hazards analyses for mortality showed significant hazard ratios for age, peak AST, and FIB-4 index on admission (p = 0.0001 and 0.0108, respectively), but not in a model including AST and FIB-4 index on admission. Furthermore, the AST peak was significantly higher among non-surviving patients with DM than in those without DM. CONCLUSIONS: We found that advanced age, high AST, and FIB-4 index on admission and a higher peak AST during hospitalization are risk factors for poor COVID-19 prognosis. Furthermore, DM was a risk factor for exacerbation of liver injury among non-surviving patients. The AST concentration and FIB-4 index should be assessed periodically throughout hospitalization, especially in patients with high AST values on admission and those with DM.

5.
Nat Commun ; 11(1): 4660, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32938908

ABSTRACT

Intratumor spatial heterogeneity facilitates therapeutic resistance in glioblastoma (GBM). Nonetheless, understanding of GBM heterogeneity is largely limited to the surgically resectable tumor core lesion while the seeds for recurrence reside in the unresectable tumor edge. In this study, stratification of GBM to core and edge demonstrates clinically relevant surgical sequelae. We establish regionally derived models of GBM edge and core that retain their spatial identity in a cell autonomous manner. Upon xenotransplantation, edge-derived cells show a higher capacity for infiltrative growth, while core cells demonstrate core lesions with greater therapy resistance. Investigation of intercellular signaling between these two tumor populations uncovers the paracrine crosstalk from tumor core that promotes malignancy and therapy resistance of edge cells. These phenotypic alterations are initiated by HDAC1 in GBM core cells which subsequently affect edge cells by secreting the soluble form of CD109 protein. Our data reveal the role of intracellular communication between regionally different populations of GBM cells in tumor recurrence.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/pathology , Glioblastoma/pathology , Histone Deacetylase 1/metabolism , Neoplasm Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Female , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/mortality , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Humans , Mice, SCID , Phenylbutyrates/pharmacology , Signal Transduction , Tumor Microenvironment , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 19(5): 1134-1147, 2020 05.
Article in English | MEDLINE | ID: mdl-32127468

ABSTRACT

The development of efficacious therapies targeting metastatic spread of breast cancer to the brain represents an unmet clinical need. Accordingly, an improved understanding of the molecular underpinnings of central nervous system spread and progression of breast cancer brain metastases (BCBM) is required. In this study, the clinical burden of disease in BCBM was investigated, as well as the role of aldehyde dehydrogenase 1A3 (ALDH1A3) in the metastatic cascade leading to BCBM development. Initial analysis of clinical survival trends for breast cancer and BCBM determined improvement of breast cancer survival rates; however, this has failed to positively affect the prognostic milestones of triple-negative breast cancer (TNBC) brain metastases (BM). ALDH1A3 and a representative epithelial-mesenchymal transition (EMT) gene signature (mesenchymal markers, CD44 or Vimentin) were compared in tumors derived from BM, lung metastases (LM), or bone metastases (BoM) of patients as well as mice after injection of TNBC cells. Selective elevation of the EMT signature and ALDH1A3 were observed in BM, unlike LM and BoM, especially in the tumor edge. Furthermore, ALDH1A3 was determined to play a role in BCBM establishment via regulation of circulating tumor cell adhesion and migration phases in the BCBM cascade. Validation through genetic and pharmacologic inhibition of ALDH1A3 via lentiviral shRNA knockdown and a novel small-molecule inhibitor demonstrated selective inhibition of BCBM formation with prolonged survival of tumor-bearing mice. Given the survival benefits via targeting ALDH1A3, it may prove an effective therapeutic strategy for BCBM prevention and/or treatment.


Subject(s)
Aldehyde Oxidoreductases/antagonists & inhibitors , Brain Neoplasms/drug therapy , Gene Expression Regulation, Enzymologic/drug effects , Neoplastic Cells, Circulating/drug effects , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/enzymology , Bone Neoplasms/secondary , Brain Neoplasms/enzymology , Brain Neoplasms/secondary , Cell Proliferation , Enzyme Inhibitors/pharmacology , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Mice , Mice, SCID , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Cell Rep ; 26(7): 1893-1905.e7, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30759398

ABSTRACT

Unresectable glioblastoma (GBM) cells in the invading tumor edge can act as seeds for recurrence. The molecular and phenotypic properties of these cells remain elusive. Here, we report that the invading edge and tumor core have two distinct types of glioma stem-like cells (GSCs) that resemble proneural (PN) and mesenchymal (MES) subtypes, respectively. Upon exposure to ionizing radiation (IR), GSCs, initially enriched for a CD133+ PN signature, transition to a CD109+ MES subtype in a C/EBP-ß-dependent manner. Our gene expression analysis of paired cohorts of patients with primary and recurrent GBMs identified a CD133-to-CD109 shift in tumors with an MES recurrence. Patient-derived CD133-/CD109+ cells are highly enriched with clonogenic, tumor-initiating, and radiation-resistant properties, and silencing CD109 significantly inhibits these phenotypes. We also report a conserved regulation of YAP/TAZ pathways by CD109 that could be a therapeutic target in GBM.


Subject(s)
Adaptation, Physiological/genetics , Glioma/radiotherapy , Radiation, Ionizing , Glioma/pathology , Humans
9.
Cancer Cell ; 34(1): 119-135.e10, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29937354

ABSTRACT

Aggressive cancers such as glioblastoma (GBM) contain intermingled apoptotic cells adjacent to proliferating tumor cells. Nonetheless, intercellular signaling between apoptotic and surviving cancer cells remain elusive. In this study, we demonstrate that apoptotic GBM cells paradoxically promote proliferation and therapy resistance of surviving tumor cells by secreting apoptotic extracellular vesicles (apoEVs) enriched with various components of spliceosomes. apoEVs alter RNA splicing in recipient cells, thereby promoting their therapy resistance and aggressive migratory phenotype. Mechanistically, we identified RBM11 as a representative splicing factor that is upregulated in tumors after therapy and shed in extracellular vesicles upon induction of apoptosis. Once internalized in recipient cells, exogenous RBM11 switches splicing of MDM4 and Cyclin D1 toward the expression of more oncogenic isoforms.


Subject(s)
Apoptosis , Brain Neoplasms/metabolism , Extracellular Vesicles/metabolism , Glioblastoma/metabolism , RNA-Binding Proteins/metabolism , Spliceosomes/metabolism , Animals , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Communication , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Drug Resistance, Neoplasm , Extracellular Vesicles/drug effects , Extracellular Vesicles/genetics , Extracellular Vesicles/pathology , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Splicing , RNA-Binding Proteins/genetics , Signal Transduction , Spliceosomes/drug effects , Spliceosomes/genetics , Spliceosomes/pathology , Tumor Burden
10.
Cancer Res ; 78(11): 3002-3013, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29531161

ABSTRACT

Glioblastoma (GBM) is a lethal disease with no effective therapies available. We previously observed upregulation of the TAM (Tyro-3, Axl, and Mer) receptor tyrosine kinase family member AXL in mesenchymal GBM and showed that knockdown of AXL induced apoptosis of mesenchymal, but not proneural, glioma sphere cultures (GSC). In this study, we report that BGB324, a novel small molecule inhibitor of AXL, prolongs the survival of immunocompromised mice bearing GSC-derived mesenchymal GBM-like tumors. We show that protein S (PROS1), a known ligand of other TAM receptors, was secreted by tumor-associated macrophages/microglia and subsequently physically associated with and activated AXL in mesenchymal GSC. PROS1-driven phosphorylation of AXL (pAXL) induced NFκB activation in mesenchymal GSC, which was inhibited by BGB324 treatment. We also found that treatment of GSC-derived mouse GBM tumors with nivolumab, a blocking antibody against the immune checkpoint protein PD-1, increased intratumoral macrophages/microglia and activation of AXL. Combinatorial therapy with nivolumab plus BGB324 effectively prolonged the survival of mice bearing GBM tumors. Clinically, expression of AXL or PROS1 was associated with poor prognosis for patients with GBM. Our results suggest that the PROS1-AXL pathway regulates intrinsic mesenchymal signaling and the extrinsic immune microenvironment, contributing to the growth of aggressive GBM tumors.Significance: These findings suggest that development of combination treatments of AXL and immune checkpoint inhibitors may provide benefit to patients with GBM. Cancer Res; 78(11); 3002-13. ©2018 AACR.


Subject(s)
Glioblastoma/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Microenvironment/physiology , Animals , Apoptosis/physiology , Benzocycloheptenes/pharmacology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Glioblastoma/drug therapy , Glioma/metabolism , Humans , Male , Mice , Middle Aged , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Triazoles/pharmacology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
11.
J Clin Invest ; 127(8): 3075-3089, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28737508

ABSTRACT

Accumulating evidence suggests that glioma stem cells (GSCs) are important therapeutic targets in glioblastoma (GBM). In this study, we identified NIMA-related kinase 2 (NEK2) as a functional binding protein of enhancer of zeste homolog 2 (EZH2) that plays a critical role in the posttranslational regulation of EZH2 protein in GSCs. NEK2 was among the most differentially expressed kinase-encoding genes in GSC-containing cultures (glioma spheres), and it was required for in vitro clonogenicity, in vivo tumor propagation, and radioresistance. Mechanistically, the formation of a protein complex comprising NEK2 and EZH2 in glioma spheres phosphorylated and then protected EZH2 from ubiquitination-dependent protein degradation in a NEK2 kinase activity-dependent manner. Clinically, NEK2 expression in patients with glioma was closely associated with EZH2 expression and correlated with a poor prognosis. NEK2 expression was also substantially elevated in recurrent tumors after therapeutic failure compared with primary untreated tumors in matched GBM patients. We designed a NEK2 kinase inhibitor, compound 3a (CMP3a), which efficiently attenuated GBM growth in a mouse model and exhibited a synergistic effect with radiotherapy. These data demonstrate a key role for NEK2 in maintaining GSCs in GBM by stabilizing the EZH2 protein and introduce the small-molecule inhibitor CMP3a as a potential therapeutic agent for GBM.


Subject(s)
Brain Neoplasms/drug therapy , Enhancer of Zeste Homolog 2 Protein/metabolism , Glioblastoma/drug therapy , NIMA-Related Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/radiotherapy , Female , Gene Silencing , Glioblastoma/radiotherapy , Humans , Mice , Mice, Nude , NIMA-Related Kinases/chemistry , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Phosphorylation
12.
Cancer Res ; 76(24): 7219-7230, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27569208

ABSTRACT

Glioma stem-like cells (GSC) with tumor-initiating activity orchestrate the cellular hierarchy in glioblastoma and engender therapeutic resistance. Recent work has divided GSC into two subtypes with a mesenchymal (MES) GSC population as the more malignant subtype. In this study, we identify the FOXD1-ALDH1A3 signaling axis as a determinant of the MES GSC phenotype. The transcription factor FOXD1 is expressed predominantly in patient-derived cultures enriched with MES, but not with the proneural GSC subtype. shRNA-mediated attenuation of FOXD1 in MES GSC ablates their clonogenicity in vitro and in vivo Mechanistically, FOXD1 regulates the transcriptional activity of ALDH1A3, an established functional marker for MES GSC. Indeed, the functional roles of FOXD1 and ALDH1A3 are likely evolutionally conserved, insofar as RNAi-mediated attenuation of their orthologous genes in Drosophila blocks formation of brain tumors engineered in that species. In clinical specimens of high-grade glioma, the levels of expression of both FOXD1 and ALDH1A3 are inversely correlated with patient prognosis. Finally, a novel small-molecule inhibitor of ALDH we developed, termed GA11, displays potent in vivo efficacy when administered systemically in a murine GSC-derived xenograft model of glioblastoma. Collectively, our findings define a FOXD1-ALDH1A3 pathway in controling the clonogenic and tumorigenic potential of MES GSC in glioblastoma tumors. Cancer Res; 76(24); 7219-30. ©2016 AACR.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Brain Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Glioma/pathology , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Brain Neoplasms/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drosophila , Fluorescent Antibody Technique , Glioma/metabolism , Humans , Immunohistochemistry , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Nude , Microscopy, Confocal , Signal Transduction/physiology , Xenograft Model Antitumor Assays
13.
Nano Lett ; 16(9): 5326-32, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27420544

ABSTRACT

Enhanced glioma-stem-cell (GSC) motility and therapy resistance are considered to play key roles in tumor cell dissemination and recurrence. As such, a better understanding of the mechanisms by which these cells disseminate and withstand therapy could lead to more efficacious treatments. Here, we introduce a novel micro-/nanotechnology-enabled chip platform for performing live-cell interrogation of patient-derived GSCs with single-clone resolution. On-chip analysis revealed marked intertumoral differences (>10-fold) in single-clone motility profiles between two populations of GSCs, which correlated well with results from tumor-xenograft experiments and gene-expression analyses. Further chip-based examination of the more-aggressive GSC population revealed pronounced interclonal variations in motility capabilities (up to ∼4-fold) as well as gene-expression profiles at the single-cell level. Chip-supported therapy resistance studies with a chemotherapeutic agent (i.e., temozolomide) and an oligo RNA (anti-miR363) revealed a subpopulation of CD44-high GSCs with strong antiapoptotic behavior as well as enhanced motility capabilities. The living-cell-interrogation chip platform described herein enables thorough and large-scale live monitoring of heterogeneous cancer-cell populations with single-cell resolution, which is not achievable by any other existing technology and thus has the potential to provide new insights into the cellular and molecular mechanisms modulating glioma-stem-cell dissemination and therapy resistance.


Subject(s)
Brain Neoplasms/pathology , Cell Movement , Glioblastoma/pathology , Neoplastic Stem Cells/cytology , Animals , Apoptosis , Humans , Mice , Tumor Cells, Cultured
14.
Genom Data ; 5: 333-336, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26251826

ABSTRACT

Tumor heterogeneity of high-grade glioma (HGG) is recognized by four clinically relevant subtypes based on core gene signatures. However, molecular signaling in glioma stem cells (GSCs) in individual HGG subtypes is poorly characterized. Previously we identified and characterized two mutually exclusive GSC subtypes with distinct activated signaling pathways and biological phenotypes. One GSC subtype presented with a gene signature resembling Proneural (PN) HGG, whereas the other was similar to Mesenchymal (Mes) HGG. Classical HGG-derived GSCs were sub-classified as either one of these two subtypes. Differential mRNA expression analysis of PN and Mes GSCs identified 5,796 differentially expressed genes, revealing a pronounced correlation with the corresponding PN or Mes HGGs. Mes GSCs displayed more aggressive phenotypes in vitro and as intracranial xenografts in mice. Further, Mes GSCs were markedly resistant to radiation compared with PN GSCs. Expression of ALDH1A3 - one of the most up-regulated Mes representative genes and a universal cancer stem cell marker in non-brain cancers - was associated with self-renewal and a multi-potent stem cell population in Mes but not PN samples. Moreover, inhibition of ALDH1A3 attenuated the growth of Mes but not PN GSCs in vitro. Lastly, radiation treatment of PN GSCs up-regulated Mes-associated markers and down-regulated PN-associated markers, whereas inhibition of ALDH1A3 attenuated an irradiation-induced gain of Mes identity in PN GSCs in vitro. Taken together, our data suggest that two subtypes of GSCs, harboring distinct metabolic signaling pathways, represent intertumoral glioma heterogeneity and highlight previously unidentified roles of ALDH1A3-associated signaling that promotes aberrant proliferation of Mes HGGs and GSCs. Inhibition of ALDH1A3-mediated pathways therefore might provide a promising therapeutic approach for a subset of HGGs with the Mes signature. Here, we describe the gene expression analysis, including pre-processing methods for the data published by Mao and colleagues in PNAS [1], integration of microarray data from this study with The Cancer Genome Atlas (TCGA) glioblastoma data and also with another published study.

15.
Springerplus ; 4: 41, 2015.
Article in English | MEDLINE | ID: mdl-25694859

ABSTRACT

INTRODUCTION: TNF-α inhibitors plus MTX appear to have benefit in the longer-term reduction of RA. Boolean long-term remission under drug-free conditions is rare. The therapeutic mechanism and the factor of predicting response have not been clarified yet. CASE DESCRIPTION: A 24-year-old female rheumatoid arthritis (RA) patient, who once attained complete remission (CR) with the combination therapy with tumor necrosis factor alpha (TNF-alpha) inhibitor adalimumab (ADA) and methotrexate (MTX), showed the occurrence of Epstain- Barr virus (EBV)-associated lymphoproliferative disorder (LPD). Pulse treatment with methylprednisolone after the termination of anti TNF-α therapy resulted in the remission of EBV-associated LPD. The administration of prednisolone (PSL) was tapered off after the improvement of clinical symptoms and laboratory data. The patients achieved drug-free 12 months after urgent hospitalization and delivered healthy baby 2 years after hospital discharge. She has been complete drug-free Boolean remission for 5 years. DISCUSSION AND EVALUATION: The purpose of this brief case is report that we experienced the remission of LPD after CR with combined therapy with ADA and MTX. We believe this case report will be one of the paths for unveiling the pathogenesis and improving the treatment for RA. CONCLUSIONS: We believe this case report will be one of the paths for unveiling the pathogenesis and improving the treatment for RA.

16.
Semin Cell Dev Biol ; 40: 17-26, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25721810

ABSTRACT

Extracellular vesicles (EVs) act as carriers of molecular and oncogenic signatures present in subsets of tumour cells and tumour-associated stroma, and as mediators of intercellular communication. These processes likely involve cancer stem cells (CSCs). EVs represent a unique pathway of cellular export and cell-to-cell transfer of insoluble molecular regulators such as membrane receptors, signalling proteins and metabolites, thereby influencing the functional integration of cancer cell populations. While mechanisms that control biogenesis, cargo and uptake of different classes of EVs (exosomes, microvesicles, ectosomes, large oncosomes) are poorly understood, they likely remain under the influence of stress-responses, microenvironment and oncogenic processes that define the biology and heterogeneity of human cancers. In glioblastoma (GBM), recent molecular profiling approaches distinguished several disease subtypes driven by distinct molecular, epigenetic and mutational mechanisms, leading to formation of proneural, neural, classical and mesenchymal tumours. Moreover, molecularly distinct clonal cellular lineages co-exist within individual GBM lesions, where they differentiate according to distinct stem cell hierarchies resulting in several facets of tumour heterogeneity and the related potential for intercellular interactions. Glioma stem cells (GSCs) may carry signatures of either proneural or mesenchymal GBM subtypes and differ in several biological characteristics that are, at least in part, represented by the output and repertoire of EV production (vesiculome). We report that vesiculomes differ between known GBM subtypes. EVs may also reflect and influence the equilibrium of the stem cell hierarchy, contain oncogenic drivers and modulate the microenvironment (vascular niche). The GBM/GSC subtype-specific differentials in EV cargo of proteins, transcripts, microRNA and DNA may enable detection of the dynamics of the stem cell compartment and result in biological effects that remain to be fully characterized.


Subject(s)
Extracellular Vesicles/pathology , Animals , Biomarkers/chemistry , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Communication , Extracellular Vesicles/chemistry , Extracellular Vesicles/drug effects , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/pathology
17.
Stem Cell Reports ; 4(2): 226-38, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25601206

ABSTRACT

Glioblastoma (GBM)-derived tumorigenic stem-like cells (GSCs) may play a key role in therapy resistance. Previously, we reported that the mitotic kinase MELK binds and phosphorylates the oncogenic transcription factor FOXM1 in GSCs. Here, we demonstrate that the catalytic subunit of Polycomb repressive complex 2, EZH2, is targeted by the MELK-FOXM1 complex, which in turn promotes resistance to radiation in GSCs. Clinically, EZH2 and MELK are coexpressed in GBM and significantly induced in postirradiation recurrent tumors whose expression is inversely correlated with patient prognosis. Through a gain-and loss-of-function study, we show that MELK or FOXM1 contributes to GSC radioresistance by regulation of EZH2. We further demonstrate that the MELK-EZH2 axis is evolutionarily conserved in Caenorhabditis elegans. Collectively, these data suggest that the MELK-FOXM1-EZH2 signaling axis is essential for GSC radioresistance and therefore raise the possibility that MELK-FOXM1-driven EZH2 signaling can serve as a therapeutic target in irradiation-resistant GBM tumors.


Subject(s)
Glioma/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Polycomb Repressive Complex 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Death/genetics , Cell Death/radiation effects , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein , Gene Expression , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/mortality , Heterografts , Humans , Mice , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Transport , Radiation Tolerance/genetics , Signal Transduction , Transcription, Genetic
18.
Stem Cells Transl Med ; 3(11): 1262-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25232184

ABSTRACT

Glioblastoma (GBM) is a primary brain cancer with an extremely poor prognosis. GBM tumors contain heterogeneous cellular components, including a small subpopulation of tumor cells termed glioma stem cells (GSCs). GSCs are characterized as chemotherapy- and radiotherapy-resistant cells with prominent tumorigenic ability. Studies in Drosophila cancer models demonstrated that interclonal cooperation and signaling from apoptotic clones provokes aggressive growth of neighboring tumorigenic clones, via compensatory proliferation or apoptosis induced proliferation. Mechanistically, these aggressive tumors depend on activation of Jun-N-terminal kinase (upstream of c-JUN), and Drosophila Wnt (Wg) in the apoptotic clones. Consistent with these nonmammalian studies, data from several mammalian studies have shown that c-JUN and Wnt are hyperactivated in aggressive tumors (including GBM). However, it remains elusive whether compensatory proliferation is an evolutionarily conserved mechanism in cancers. In the present report, we summarize recent studies in Drosophila models and mammalian models (e.g., xenografts of human cancer cells into small animals) to elucidate the intercellular interactions between the apoptosis-prone cancer cells (e.g., non-GSCs) and the hyperproliferative cancer cells (e.g., GSCs). These evolving investigations will yield insights about molecular signaling interactions in the context of post-therapeutic phenotypic changes in human cancers. Furthermore, these studies are likely to revise our understanding of the genetic changes and post-therapeutic cell-cell interactions, which is a vital area of cancer biology with wide applications to many cancer types in humans.


Subject(s)
Brain Neoplasms , Glioblastoma , Neoplasms, Experimental , Neoplastic Stem Cells , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Drosophila Proteins/metabolism , Drosophila melanogaster , Drug Resistance, Neoplasm , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Radiation Tolerance , Wnt Proteins/metabolism
19.
J Clin Invest ; 124(9): 3741-56, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25061874

ABSTRACT

Aberrant activation of EGFR in human cancers promotes tumorigenesis through stimulation of AKT signaling. Here, we determined that the discoidina neuropilin-like membrane protein DCBLD2 is upregulated in clinical specimens of glioblastomas and head and neck cancers (HNCs) and is required for EGFR-stimulated tumorigenesis. In multiple cancer cell lines, EGFR activated phosphorylation of tyrosine 750 (Y750) of DCBLD2, which is located within a recently identified binding motif for TNF receptor-associated factor 6 (TRAF6). Consequently, phosphorylation of DCBLD2 Y750 recruited TRAF6, leading to increased TRAF6 E3 ubiquitin ligase activity and subsequent activation of AKT, thereby enhancing EGFR-driven tumorigenesis. Moreover, evaluation of patient samples of gliomas and HNCs revealed an association among EGFR activation, DCBLD2 phosphorylation, and poor prognoses. Together, our findings uncover a pathway in which DCBLD2 functions as a signal relay for oncogenic EGFR signaling to promote tumorigenesis and suggest DCBLD2 and TRAF6 as potential therapeutic targets for human cancers that are associated with EGFR activation.


Subject(s)
Carcinogenesis , ErbB Receptors/physiology , Membrane Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , TNF Receptor-Associated Factor 6/metabolism , Brain Neoplasms/etiology , Cells, Cultured , Glioma/etiology , Head and Neck Neoplasms/etiology , Humans , Membrane Proteins/genetics , Phosphorylation , Signal Transduction
20.
PLoS One ; 9(4): e92546, 2014.
Article in English | MEDLINE | ID: mdl-24739874

ABSTRACT

Glioblastoma multiforme (GBM) is a highly lethal brain tumor. Due to resistance to current therapies, patient prognosis remains poor and development of novel and effective GBM therapy is crucial. Glioma stem cells (GSCs) have gained attention as a therapeutic target in GBM due to their relative resistance to current therapies and potent tumor-initiating ability. Previously, we identified that the mitotic kinase maternal embryonic leucine-zipper kinase (MELK) is highly expressed in GBM tissues, specifically in GSCs, and its expression is inversely correlated with the post-surgical survival period of GBM patients. In addition, patient-derived GSCs depend on MELK for their survival and growth both in vitro and in vivo. Here, we demonstrate evidence that the role of MELK in the GSC survival is specifically dependent on its kinase activity. With in silico structure-based analysis for protein-compound interaction, we identified the small molecule Compound 1 (C1) is predicted to bind to the kinase-active site of MELK protein. Elimination of MELK kinase activity was confirmed by in vitro kinase assay in nano-molar concentrations. When patient-derived GSCs were treated with C1, they underwent mitotic arrest and subsequent cellular apoptosis in vitro, a phenotype identical to that observed with shRNA-mediated MELK knockdown. In addition, C1 treatment strongly induced tumor cell apoptosis in slice cultures of GBM surgical specimens and attenuated growth of mouse intracranial tumors derived from GSCs in a dose-dependent manner. Lastly, C1 treatment sensitizes GSCs to radiation treatment. Collectively, these data indicate that targeting MELK kinase activity is a promising approach to attenuate GBM growth by eliminating GSCs in tumors.


Subject(s)
Complement C1 Inhibitor Protein/pharmacology , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Survival , DNA Damage , DNA Repair , Down-Regulation , Heterografts/drug effects , Heterografts/metabolism , Humans , Immunocompromised Host , Immunohistochemistry , Mice , Models, Molecular , Peptides/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL