Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Cell ; 175(5): 1228-1243.e20, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30392959

ABSTRACT

Genetic drivers of cancer can be dysregulated through epigenetic modifications of DNA. Although the critical role of DNA 5-methylcytosine (5mC) in the regulation of transcription is recognized, the functions of other non-canonical DNA modifications remain obscure. Here, we report the identification of novel N6-methyladenine (N6-mA) DNA modifications in human tissues and implicate this epigenetic mark in human disease, specifically the highly malignant brain cancer glioblastoma. Glioblastoma markedly upregulated N6-mA levels, which co-localized with heterochromatic histone modifications, predominantly H3K9me3. N6-mA levels were dynamically regulated by the DNA demethylase ALKBH1, depletion of which led to transcriptional silencing of oncogenic pathways through decreasing chromatin accessibility. Targeting the N6-mA regulator ALKBH1 in patient-derived human glioblastoma models inhibited tumor cell proliferation and extended the survival of tumor-bearing mice, supporting this novel DNA modification as a potential therapeutic target for glioblastoma. Collectively, our results uncover a novel epigenetic node in cancer through the DNA modification N6-mA.


Subject(s)
Adenine/analogs & derivatives , Brain Neoplasms/pathology , DNA Methylation , Glioblastoma/pathology , Adenine/analysis , Adenine/chemistry , Adult , Aged , AlkB Homolog 1, Histone H2a Dioxygenase/antagonists & inhibitors , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Cell Hypoxia , Child , Epigenomics , Female , Glioblastoma/metabolism , Glioblastoma/mortality , Heterochromatin/metabolism , Histones/metabolism , Humans , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism
2.
Mol Cell ; 83(23): 4334-4351.e7, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37979586

ABSTRACT

Growth factor receptors rank among the most important oncogenic pathways, but pharmacologic inhibitors often demonstrate limited benefit as monotherapy. Here, we show that epidermal growth factor receptor (EGFR) signaling repressed N6-methyladenosine (m6A) levels in glioblastoma stem cells (GSCs), whereas genetic or pharmacologic EGFR targeting elevated m6A levels. Activated EGFR induced non-receptor tyrosine kinase SRC to phosphorylate the m6A demethylase, AlkB homolog 5 (ALKBH5), thereby inhibiting chromosomal maintenance 1 (CRM1)-mediated nuclear export of ALKBH5 to permit sustained mRNA m6A demethylation in the nucleus. ALKBH5 critically regulated ferroptosis through m6A modulation and YTH N6-methyladenosine RNA binding protein (YTHDF2)-mediated decay of the glutamate-cysteine ligase modifier subunit (GCLM). Pharmacologic targeting of ALKBH5 augmented the anti-tumor efficacy of EGFR and GCLM inhibitors, supporting an EGFR-ALKBH5-GCLM oncogenic axis. Collectively, EGFR reprograms the epitranscriptomic landscape through nuclear retention of the ALKBH5 demethylase to protect against ferroptosis, offering therapeutic paradigms for the treatment of lethal cancers.


Subject(s)
AlkB Homolog 5, RNA Demethylase , ErbB Receptors , Ferroptosis , Glioblastoma , Humans , Adenosine/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , ErbB Receptors/genetics , Ferroptosis/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , RNA, Messenger/genetics
3.
Cell ; 153(1): 139-52, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23540695

ABSTRACT

Glioblastomas (GBMs) are highly vascular and lethal brain tumors that display cellular hierarchies containing self-renewing tumorigenic glioma stem cells (GSCs). Because GSCs often reside in perivascular niches and may undergo mesenchymal differentiation, we interrogated GSC potential to generate vascular pericytes. Here, we show that GSCs give rise to pericytes to support vessel function and tumor growth. In vivo cell lineage tracing with constitutive and lineage-specific fluorescent reporters demonstrated that GSCs generate the majority of vascular pericytes. Selective elimination of GSC-derived pericytes disrupts the neovasculature and potently inhibits tumor growth. Analysis of human GBM specimens showed that most pericytes are derived from neoplastic cells. GSCs are recruited toward endothelial cells via the SDF-1/CXCR4 axis and are induced to become pericytes predominantly by transforming growth factor ß. Thus, GSCs contribute to vascular pericytes that may actively remodel perivascular niches. Therapeutic targeting of GSC-derived pericytes may effectively block tumor progression and improve antiangiogenic therapy.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Pericytes/pathology , Animals , Brain/pathology , Brain Neoplasms/blood supply , Cell Differentiation , Endothelial Cells/pathology , Glioblastoma/blood supply , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Transforming Growth Factor beta/metabolism , Transplantation, Heterologous
4.
EMBO J ; 41(7): e109187, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35191554

ABSTRACT

Hypoxia regulates tumor angiogenesis, metabolism, and therapeutic response in malignant cancers including glioblastoma, the most lethal primary brain tumor. The regulation of HIF transcriptional factors by the ubiquitin-proteasome system is critical in the hypoxia response, but hypoxia-inducible deubiquitinases that counteract the ubiquitination remain poorly defined. While the activation of ERK1/2 also plays an important role in hypoxia response, the relationship between ERK1/2 activation and HIF regulation remains elusive. Here, we identified USP33 as essential deubiquitinase that stabilizes HIF-2alpha protein in an ERK1/2-dependent manner to promote hypoxia response in cancer cells. USP33 is preferentially induced in glioma stem cells by hypoxia and interacts with HIF-2alpha, leading to its stabilization through deubiquitination. The activation of ERK1/2 upon hypoxia promoted HIF-2alpha phosphorylation, enhancing its interaction with USP33. Silencing of USP33 disrupted glioma stem cells maintenance, reduced tumor vascularization, and inhibited glioblastoma growth. Our findings highlight USP33 as an essential regulator of hypoxia response in cancer stem cells, indicating a novel potential therapeutic target for brain tumor treatment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Brain Neoplasms , Glioma , Neoplastic Stem Cells , Ubiquitin Thiolesterase , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/pathology , Cell Hypoxia , Glioma/pathology , Humans , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
5.
Stem Cells ; 41(8): 762-774, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37280108

ABSTRACT

Glioblastoma stem cells (GSCs) have unique properties of self-renewal and tumor initiation that make them potential therapeutic targets. Development of effective therapeutic strategies against GSCs requires both specificity of targeting and intracranial penetration through the blood-brain barrier. We have previously demonstrated the use of in vitro and in vivo phage display biopanning strategies to isolate glioblastoma targeting peptides. Here we selected a 7-amino acid peptide, AWEFYFP, which was independently isolated in both the in vitro and in vivo screens and demonstrated that it was able to target GSCs over differentiated glioma cells and non-neoplastic brain cells. When conjugated to Cyanine 5.5 and intravenously injected into mice with intracranially xenografted glioblastoma, the peptide localized to the site of the tumor, demonstrating intracranial tumor targeting specificity. Immunoprecipitation of the peptide with GSC proteins revealed Cadherin 2 as the glioblastoma cell surface receptor targeted by the peptides. Peptide targeting of Cadherin 2 on GSCs was confirmed through ELISA and in vitro binding analysis. Interrogation of glioblastoma databases demonstrated that Cadherin 2 expression correlated with tumor grade and survival. These results confirm that phage display can be used to isolate unique tumor-targeting peptides specific for glioblastoma. Furthermore, analysis of these cell specific peptides can lead to the discovery of cell specific receptor targets that may serve as the focus of future theragnostic tumor-homing modalities for the development of precision strategies for the treatment and diagnosis of glioblastomas.


Subject(s)
Cadherins , Cell Surface Display Techniques , Glioblastoma , Peptides , Glioblastoma/drug therapy , Glioblastoma/pathology , Neoplastic Stem Cells , Humans , Animals , Mice , Neoplasm Transplantation , Peptides/therapeutic use , Cadherins/antagonists & inhibitors , Molecular Targeted Therapy , Disease Models, Animal
6.
Stem Cells ; 39(7): 853-865, 2021 07.
Article in English | MEDLINE | ID: mdl-33594762

ABSTRACT

Glioblastoma (GBM) ranks among the most lethal of human malignancies with GBM stem cells (GSCs) that contribute to tumor growth and therapeutic resistance. Identification and isolation of GSCs continue to be a challenge, as definitive methods to purify these cells for study or targeting are lacking. Here, we leveraged orthogonal in vitro and in vivo phage display biopanning strategies to isolate a single peptide with GSC-specific binding properties. In silico analysis of this peptide led to the isolation of EYA1 (Eyes Absent 1), a tyrosine phosphatase and transcriptional coactivator. Validating the phage discovery methods, EYA1 was preferentially expressed in GSCs compared to differentiated tumor progeny. MYC is a central mediator of GSC maintenance but has been resistant to direct targeting strategies. Based on correlation and colocalization of EYA1 and MYC, we interrogated a possible interaction, revealing binding of EYA1 to MYC and loss of MYC expression upon targeting EYA1. Supporting a functional role for EYA1, targeting EYA1 expression decreased GSC proliferation, migration, and self-renewal in vitro and tumor growth in vivo. Collectively, our results suggest that phage display can identify novel therapeutic targets in stem-like tumor cells and that an EYA1-MYC axis represents a potential therapeutic paradigm for GBM.


Subject(s)
Bacteriophages , Brain Neoplasms , Glioblastoma , Bacteriophages/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Glioblastoma/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Neoplastic Stem Cells/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism
7.
Genes Dev ; 26(11): 1247-62, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22661233

ABSTRACT

Growth factor-mediated proliferation and self-renewal maintain tissue-specific stem cells and are frequently dysregulated in cancers. Platelet-derived growth factor (PDGF) ligands and receptors (PDGFRs) are commonly overexpressed in gliomas and initiate tumors, as proven in genetically engineered models. While PDGFRα alterations inform intertumoral heterogeneity toward a proneural glioblastoma (GBM) subtype, we interrogated the role of PDGFRs in intratumoral GBM heterogeneity. We found that PDGFRα is expressed only in a subset of GBMs, while PDGFRß is more commonly expressed in tumors but is preferentially expressed by self-renewing tumorigenic GBM stem cells (GSCs). Genetic or pharmacological targeting of PDGFRß (but not PDGFRα) attenuated GSC self-renewal, survival, tumor growth, and invasion. PDGFRß inhibition decreased activation of the cancer stem cell signaling node STAT3, while constitutively active STAT3 rescued the loss of GSC self-renewal caused by PDGFRß targeting. In silico survival analysis demonstrated that PDGFRB informed poor prognosis, while PDGFRA was a positive prognostic factor. Our results may explain mixed clinical responses of anti-PDGFR-based approaches and suggest the need for integration of models of cancer as an organ system into development of cancer therapies.


Subject(s)
Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Cell Proliferation , Cell Survival , Gene Knockdown Techniques , Glioblastoma/metabolism , Humans , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , STAT3 Transcription Factor/metabolism , Transplantation, Heterologous
8.
Lab Invest ; 98(7): 924-934, 2018 07.
Article in English | MEDLINE | ID: mdl-29765109

ABSTRACT

The microvascular profile has been included in the WHO glioma grading criteria. Nevertheless, microvessels in gliomas of the same WHO grade, e.g., WHO IV glioblastoma (GBM), exhibit heterogeneous and polymorphic morphology, whose possible clinical significance remains to be determined. In this study, we employed a fractal geometry-derived parameter, microvascular fractal dimension (mvFD), to quantify microvessel complexity and developed a home-made macro in Image J software to automatically determine mvFD from the microvessel-stained immunohistochemical images of GBM. We found that mvFD effectively quantified the morphological complexity of GBM microvasculature. Furthermore, high mvFD favored the survival of GBM patients as an independent prognostic indicator and predicted a better response to chemotherapy of GBM patients. When investigating the underlying relations between mvFD and tumor growth by deploying Ki67/mvFD as an index for microvasculature-normalized tumor proliferation, we discovered an inverse correlation between mvFD and Ki67/mvFD. Furthermore, mvFD inversely correlated with the expressions of a glycolytic marker, LDHA, which indicated poor prognosis of GBM patients. Conclusively, we developed an automatic approach for mvFD measurement, and demonstrated that mvFD could predict the prognosis and response to chemotherapy of GBM patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms , Glioma , Image Interpretation, Computer-Assisted/methods , Microvessels/pathology , Neovascularization, Pathologic/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Fractals , Glioma/diagnostic imaging , Glioma/drug therapy , Glioma/pathology , Humans , Immunohistochemistry , Microvessels/diagnostic imaging , Neoplasm Grading/methods , Neovascularization, Pathologic/diagnostic imaging , Prognosis
9.
Br J Cancer ; 119(10): 1267-1277, 2018 11.
Article in English | MEDLINE | ID: mdl-30318507

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a heterogeneous disease with distinct clinical subsets based on underlying genetic and epigenetic changes. DNA hypermethylation yields a unique CRC subset with a distinct phenotype and clinical behaviour, but this oncogenic pathway is not fully characterised. This study identifies and characterises miR-1247 as a novel tumour suppressor microRNA in methylated human colon cancers. METHOD: Tumour samples from patients with hypermethylated and non-methylated colon cancer and cell lines were evaluated for miR-1247 expression and function. A murine subcutaneous xenograft model was used for in vivo functional studies. RESULTS: miR-1247 was methylated and underexpressed in methylator colon cancers. Overexpression of miR-1247 significantly inhibited cell proliferation, decreased tumour cell motility, induced apoptosis, and mitigated tumour formation capacity both in vivo and in vitro. Pharmacologic demethylation increased miR-1247 expression and produced similar anti-tumour activities. Mechanistic investigations revealed that MYCBP2, a member of the c-myc oncogene family, is a direct functional target of miR-1247. Furthermore, in CRC patients, MYCBP2 protein levels are associated with miR-1247 levels and survival. CONCLUSIONS: miR-1247 acts as a tumour suppressor by inhibiting MYCBP2 in methylator colon cancer. The MYCBP2/c-myc axis may underlie the anti-tumour activities of miR-1247 and is a potential therapeutic target via demethylation agents.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colonic Neoplasms/genetics , DNA Methylation , Epigenesis, Genetic , Genes, Tumor Suppressor , MicroRNAs/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Heterografts , Humans , Mice , Mice, Nude
10.
EMBO J ; 30(5): 800-13, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21297581

ABSTRACT

Glioblastomas (GBMs) are highly lethal brain tumours with current therapies limited to palliation due to therapeutic resistance. We previously demonstrated that GBM stem cells (GSCs) display a preferential activation of DNA damage checkpoint and are relatively resistant to radiation. However, the molecular mechanisms underlying the preferential checkpoint response in GSCs remain undefined. Here, we show that L1CAM (CD171) regulates DNA damage checkpoint responses and radiosensitivity of GSCs through nuclear translocation of L1CAM intracellular domain (L1-ICD). Targeting L1CAM by RNA interference attenuated DNA damage checkpoint activation and repair, and sensitized GSCs to radiation. L1CAM regulates expression of NBS1, a critical component of the MRE11-RAD50-NBS1 (MRN) complex that activates ataxia telangiectasia mutated (ATM) kinase and early checkpoint response. Ectopic expression of NBS1 in GSCs rescued the decreased checkpoint activation and radioresistance caused by L1CAM knockdown, demonstrating that L1CAM signals through NBS1 to regulate DNA damage checkpoint responses. Mechanistically, nuclear translocation of L1-ICD mediates NBS1 upregulation via c-Myc. These data demonstrate that L1CAM augments DNA damage checkpoint activation and radioresistance of GSCs through L1-ICD-mediated NBS1 upregulation and the enhanced MRN-ATM-Chk2 signalling.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Nuclear Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Comet Assay , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunoblotting , Neoplastic Stem Cells/pathology , Neural Cell Adhesion Molecule L1/antagonists & inhibitors , Neural Cell Adhesion Molecule L1/genetics , Nuclear Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Radiation Tolerance , Radiation, Ionizing , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Stem Cells ; 32(8): 2033-47, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24831540

ABSTRACT

Glioblastomas are highly lethal brain tumors containing tumor-propagating glioma stem cells (GSCs). The molecular mechanisms underlying the maintenance of the GSC phenotype are not fully defined. Here we demonstrate that the zinc finger and X-linked transcription factor (ZFX) maintains GSC self-renewal and tumorigenic potential by upregulating c-Myc expression. ZFX is differentially expressed in GSCs relative to non-stem glioma cells and neural progenitor cells. Disrupting ZFX by shRNA reduced c-Myc expression and potently inhibited GSC self-renewal and tumor growth. Ectopic expression of c-Myc to its endogenous level rescued the effects caused by ZFX disruption, supporting that ZFX controls GSC properties through c-Myc. Furthermore, ZFX binds to a specific sequence (GGGCCCCG) on the human c-Myc promoter to upregulate c-Myc expression. These data demonstrate that ZFX functions as a critical upstream regulator of c-Myc and plays essential roles in the maintenance of the GSC phenotype. This study also supports that c-Myc is a dominant driver linking self-renewal to malignancy.


Subject(s)
Glioblastoma/pathology , Kruppel-Like Transcription Factors/metabolism , Neoplastic Stem Cells/pathology , Cell Proliferation/physiology , Cells, Cultured , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Glioblastoma/metabolism , Humans , Immunoblotting , Immunohistochemistry , In Situ Nick-End Labeling , Neoplastic Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
Neuro Oncol ; 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38456228

ABSTRACT

BACKGROUND: Hypoxia is associated with poor prognosis in many cancers including glioblastoma (GBM). Glioma stem-like cells (GSCs) often reside in hypoxic regions and serve as reservoirs for disease progression. Long non-coding RNAs (lncRNAs) have been implicated in GBM. However, the lncRNAs that modulate GSC adaptations to hypoxia are poorly understood. Identification of these lncRNAs may provide new therapeutic strategies to target GSCs under hypoxia. METHODS: lncRNAs induced by hypoxia in GSCs were identified by RNAseq. LUCAT1 expression was assessed by qPCR, RNAseq, Northern blot, single molecule FISH in GSCs, and interrogated in IvyGAP, TCGA, and CGGA databases. LUCAT1 was depleted by shRNA, CRISPR/Cas9, and CRISPR/Cas13d. RNAseq, Western blot, immunohistochemistry, co-IP, ChIP, ChIPseq, RNA immunoprecipitation, and proximity ligation assay were performed to investigate mechanisms of action of LUCAT1. GSC viability, limiting dilution assay, and tumorigenic potential in orthotopic GBM xenograft models were performed to assess the functional consequences of depleting LUCAT1. RESULTS: A new isoform of Lucat1 is induced by HIF1α and NRF2 in GSCs under hypoxia. LUCAT1 is highly expressed in hypoxic regions in GBM. Mechanistically, LUCAT1 formed a complex with HIF1α and its co-activator CBP to regulate HIF1α target gene expression and GSC adaptation to hypoxia. Depletion of LUCAT1 impaired GSC self-renewal. Silencing LUCAT1 decreased tumor growth and prolonged mouse survival in GBM xenograft models. CONCLUSIONS: A HIF1α-LUCAT1 axis forms a positive feedback loop to amplify HIF1α signaling in GSCs under hypoxia. LUCAT1 promotes GSC self-renewal and GBM tumor growth. LUCAT1 is a potential therapeutic target in GBM.

13.
Ann Neurol ; 72(5): 766-78, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23280793

ABSTRACT

OBJECTIVE: Glioblastomas (GBMs) are lethal cancers that display cellular hierarchies parallel to normal brain. At the apex are GBM stem cells (GSCs), which are relatively resistant to conventional therapy. Interactions with the adjacent perivascular niche are an important driver of malignancy and self-renewal in GSCs. Extracellular matrix (ECM) cues instruct neural stem/progenitor cell-niche interactions, and the objective of our study was to elucidate its composition and contribution to GSC maintenance in the perivascular niche. METHODS: We interrogated human tumor tissue for immunofluorescence analysis and derived GSCs from tumor tissues for functional studies. Bioinformatics analyses were conducted by mining publicly available databases. RESULTS: We find that laminin ECM proteins are localized to the perivascular GBM niche and inform negative patient prognosis. To identify the source of laminins, we characterized cellular elements within the niche and found that laminin α chains were expressed by nonstem tumor cells and tumor-associated endothelial cells (ECs). RNA interference targeting laminin α2 inhibited GSC growth and self-renewal. In co-culture studies of GSCs and ECs, laminin α2 knockdown in ECs resulted in decreased tumor growth. INTERPRETATION: Our studies highlight the contribution of nonstem tumor cell-derived laminin juxtracrine signaling. As laminin α2 has recently been identified as a molecular marker of aggressive ependymoma, we propose that the brain vascular ECM promotes tumor malignancy through maintenance of the GSC compartment, providing not only a molecular fingerprint but also a possible therapeutic target.


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/pathology , Laminin/metabolism , Neoplastic Stem Cells/physiology , AC133 Antigen , Analysis of Variance , Antigens, CD/metabolism , Brain Neoplasms/mortality , Cell Survival/drug effects , Coculture Techniques , Computational Biology , Dose-Response Relationship, Radiation , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/mortality , Glycoproteins/metabolism , Humans , Kaplan-Meier Estimate , Laminin/genetics , Magnetic Resonance Imaging , Male , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Peptides/metabolism , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Radiation , Regression Analysis , Time Factors , Tissue Array Analysis , Tumor Cells, Cultured , Tumor Microenvironment/physiology
14.
Cancer Cell ; 41(9): 1621-1636.e8, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37595587

ABSTRACT

Brain metastasis of lung cancer causes high mortality, but the exact mechanisms underlying the metastasis remain unclear. Here we report that vascular pericytes derived from CD44+ lung cancer stem cells (CSCs) in lung adenocarcinoma (ADC) potently cause brain metastases through the G-protein-coupled receptor 124 (GPR124)-enhanced trans-endothelial migration (TEM). CD44+ CSCs in perivascular niches generate the majority of vascular pericytes in lung ADC. CSC-derived pericyte-like cells (Cd-pericytes) exhibit remarkable TEM capacity to effectively intravasate into the vessel lumina, survive in the circulation, extravasate into the brain parenchyma, and then de-differentiate into tumorigenic CSCs to form metastases. Cd-pericytes uniquely express GPR124 that activates Wnt7-ß-catenin signaling to enhance TEM capacity of Cd-pericytes for intravasation and extravasation, two critical steps during tumor metastasis. Furthermore, selective disruption of Cd-pericytes, GPR124, or the Wnt7-ß-catenin signaling markedly reduces brain and liver metastases of lung ADC. Our findings uncover an unappreciated cellular and molecular paradigm driving tumor metastasis.


Subject(s)
Adenocarcinoma of Lung , Brain Neoplasms , Lung Neoplasms , Humans , Adenocarcinoma of Lung/secondary , beta Catenin , Brain Neoplasms/secondary , Cadmium , Hyaluronan Receptors , Lung , Lung Neoplasms/pathology , Pericytes , Receptors, G-Protein-Coupled
15.
Nat Commun ; 14(1): 2262, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37080989

ABSTRACT

The Wnt pathway is frequently dysregulated in many cancers, underscoring it as a therapeutic target. Wnt inhibitors have uniformly failed in clinical trials. Here, we report a mechanism of WNT pathway activation through the Semaphorin 3 C neurodevelopmental program in glioma stem-like cells. Sema3C directs ß-catenin nuclear accumulation in a Rac1-dependent process, leading to transactivation of Wnt target genes. Sema3C-driven Wnt signaling occurred despite suppression of Wnt ligand secretion, suggesting that Sema3C drives canonical Wnt signaling independent of Wnt ligand binding. In a mouse model of glioblastoma, combined depletion of Sema3C and ß-catenin partner TCF1 extended animal survival more than single target inhibition alone. In human glioblastoma, Sema3C expression and Wnt pathway activation were highly concordant. Since Sema3C is frequently overexpressed in glioblastoma, Sema3C signaling may be a significant mechanism of resistance to upstream Wnt pathway inhibitors. Dual targeting of Sema3C and Wnt pathways may achieve clinically significant Wnt pathway inhibition.


Subject(s)
Glioblastoma , Semaphorins , Animals , Humans , Mice , beta Catenin/genetics , beta Catenin/metabolism , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/metabolism , Ligands , Semaphorins/genetics , Wnt Signaling Pathway/genetics
16.
Neuro Oncol ; 25(8): 1428-1440, 2023 08 03.
Article in English | MEDLINE | ID: mdl-36521011

ABSTRACT

BACKGROUND: Cancer cells including cancer stem cells exhibit a higher rate of ribosome biogenesis than normal cells to support rapid cell proliferation in tumors. However, the molecular mechanisms governing the preferential ribosome biogenesis in glioma stem cells (GSCs) remain unclear. In this work, we show that the novel INHAT repressor (NIR) promotes ribosomal DNA (rDNA) transcription to support GSC proliferation and glioblastoma (GBM) growth, suggesting that NIR is a potential therapeutic target for GBM. METHODS: Immunoblotting, immunohistochemical and immunofluorescent analysis were used to determine NIR expression in GSCs and human GBMs. Using shRNA-mediated knockdown, we assessed the role and functional significance of NIR in GSCs and GSC-derived orthotopic GBM xenografts. We further performed mass spectrometry analysis, chromatin immunoprecipitation, and other biochemical assays to define the molecular mechanisms by which NIR promotes GBM progression. RESULTS: Our results show that high expression of NIR predicts poor survival in GBM patients. NIR is enriched in the nucleoli of GSCs in human GBMs. Disrupting NIR markedly suppresses GSC proliferation and tumor growth by inhibiting rDNA transcription and pre-ribosomal RNA synthesis. In mechanistic studies, we find that NIR activates rDNA transcription to promote GSC proliferation by cooperating with Nucleolin (NCL) and Nucleophosmin 1 (NPM1), 2 important nucleolar transcription factors. CONCLUSIONS: Our study uncovers a critical role of NIR-mediated rDNA transcription in the malignant progression of GBM, indicating that targeting this axis may provide a novel therapeutic strategy for GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/pathology , DNA, Ribosomal/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/pathology , Neoplastic Stem Cells/metabolism , Cell Proliferation
17.
Cancer Cell ; 5(4): 329-39, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15093540

ABSTRACT

Molecular mechanisms associated with tumor metastasis remain poorly understood. Here we report that acquired expression of periostin by colon cancer cells greatly promoted metastatic development of colon tumors. Periostin is overexpressed in more than 80% of human colon cancers examined with highest expression in metastatic tumors. Periostin expression dramatically enhanced metastatic growth of colon cancer by both preventing stress-induced apoptosis in the cancer cells and augmenting endothelial cell survival to promote angiogenesis. At the molecular level, periostin activated the Akt/PKB signaling pathway through the alpha(v)beta(3) integrins to increase cellular survival. These data demonstrated that the survival-promoting function is crucial for periostin to promote tumor metastasis of colon cancer.


Subject(s)
Cell Adhesion Molecules/metabolism , Colonic Neoplasms/pathology , Liver Neoplasms/secondary , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis , Cell Hypoxia , Cell Survival , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Integrin alphaVbeta3/metabolism , Liver Neoplasms/metabolism , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt , Signal Transduction , Tumor Cells, Cultured
18.
Nature ; 444(7120): 756-60, 2006 Dec 07.
Article in English | MEDLINE | ID: mdl-17051156

ABSTRACT

Ionizing radiation represents the most effective therapy for glioblastoma (World Health Organization grade IV glioma), one of the most lethal human malignancies, but radiotherapy remains only palliative because of radioresistance. The mechanisms underlying tumour radioresistance have remained elusive. Here we show that cancer stem cells contribute to glioma radioresistance through preferential activation of the DNA damage checkpoint response and an increase in DNA repair capacity. The fraction of tumour cells expressing CD133 (Prominin-1), a marker for both neural stem cells and brain cancer stem cells, is enriched after radiation in gliomas. In both cell culture and the brains of immunocompromised mice, CD133-expressing glioma cells survive ionizing radiation in increased proportions relative to most tumour cells, which lack CD133. CD133-expressing tumour cells isolated from both human glioma xenografts and primary patient glioblastoma specimens preferentially activate the DNA damage checkpoint in response to radiation, and repair radiation-induced DNA damage more effectively than CD133-negative tumour cells. In addition, the radioresistance of CD133-positive glioma stem cells can be reversed with a specific inhibitor of the Chk1 and Chk2 checkpoint kinases. Our results suggest that CD133-positive tumour cells represent the cellular population that confers glioma radioresistance and could be the source of tumour recurrence after radiation. Targeting DNA damage checkpoint response in cancer stem cells may overcome this radioresistance and provide a therapeutic model for malignant brain cancers.


Subject(s)
Brain Neoplasms/pathology , DNA Damage , DNA Repair , Glioma/pathology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Radiation Tolerance , AC133 Antigen , Animals , Antigens, CD/metabolism , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , DNA Damage/genetics , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/genetics , Glioma/metabolism , Glycoproteins/metabolism , Humans , Mice , Neoplastic Stem Cells/metabolism , Peptides/metabolism , Stem Cell Transplantation , Transplantation, Heterologous
19.
Sci China Life Sci ; 65(2): 362-375, 2022 02.
Article in English | MEDLINE | ID: mdl-34109474

ABSTRACT

Beef and mutton production has been aided by breeding to integrate allelic diversity for myostatin (MSTN), but a lack of diversity in the MSTN germplasm has limited similar advances in pig farming. Moreover, insurmountable challenges with congenital lameness and a dearth of data about the impacts of feed conversion, reproduction, and meat quality in MSTN-edited pigs have also currently blocked progress. Here, in a largest-to-date evaluation of multiple MSTN-edited pig populations, we demonstrated a practical alternative edit-site-based solution that overcomes the major production obstacle of hindlimb weakness. We also provide long-term and multidomain datasets for multiple breeds that illustrate how MSTN-editing can sustainably increase the yields of breed-specific lean meat and the levels of desirable lipids without deleteriously affecting feed-conversion rates or litter size. Apart from establishing a new benchmark for the data scale and quality of genome-edited animal production, our study specifically illustrates how gene-editing site selection profoundly impacts the phenotypic outcomes in diverse genetic backgrounds.


Subject(s)
Gene Editing/methods , Lameness, Animal/prevention & control , Myostatin/genetics , Pork Meat/analysis , Swine Diseases/prevention & control , Alleles , Animal Nutritional Physiological Phenomena , Animals , Animals, Genetically Modified , Energy Metabolism , Hindlimb/physiopathology , Lameness, Animal/genetics , Lameness, Animal/metabolism , Species Specificity , Swine/genetics , Swine Diseases/genetics , Swine Diseases/metabolism , Thermogenesis
20.
Sci Transl Med ; 14(626): eabf3917, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34985972

ABSTRACT

Glioblastomas are universally fatal cancers and contain self-renewing glioblastoma stem cells (GSCs) that initiate tumors. Traditional anticancer drug discovery based on in vitro cultures tends to identify targets with poor therapeutic indices and fails to accurately model the effects of the tumor microenvironment. Here, leveraging in vivo genetic screening, we identified the histone H3 lysine 4 trimethylation (H3K4me3) regulator DPY30 (Dpy-30 histone methyltransferase complex regulatory subunit) as an in vivo­specific glioblastoma dependency. On the basis of the hypothesis that in vivo epigenetic regulation may define critical GSC dependencies, we interrogated active chromatin landscapes of GSCs derived from intracranial patient-derived xenografts (PDXs) and cell culture through H3K4me3 chromatin immunoprecipitation and transcriptome analyses. Intracranial-specific genes marked by H3K4me3 included FOS, NFκB, and phosphodiesterase (PDE) family members. In intracranial PDX tumors, DPY30 regulated angiogenesis and hypoxia pathways in an H3K4me3-dependent manner but was dispensable in vitro in cultured GSCs. PDE4B was a key downstream effector of DPY30, and the PDE4 inhibitor rolipram preferentially targeted DPY30-expressing cells and impaired PDX tumor growth in mice without affecting tumor cells cultured in vitro. Collectively, the MLL/SET1 (mixed lineage leukemia/SET domain-containing 1, histone lysine methyltransferase) complex member DPY30 selectively regulates H3K4me3 modification on genes critical to support angiogenesis and tumor growth in vivo, suggesting the DPY30-PDE4B axis as a specific therapeutic target in glioblastoma.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Glioblastoma , Transcription Factors , Animals , Chromatin , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Epigenesis, Genetic , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Mice , Stem Cells/metabolism , Transcription Factors/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL