Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(45): e2307094120, 2023 Nov 07.
Article in English | MEDLINE | ID: mdl-37922327

ABSTRACT

Bone metastasis is a frequent and incurable consequence of advanced prostate cancer (PC). An interplay between disseminated tumor cells and heterogeneous bone resident cells in the metastatic niche initiates this process. Melanoma differentiation associated gene-9 (mda-9/Syntenin/syndecan binding protein) is a prometastatic gene expressed in multiple organs, including bone marrow-derived mesenchymal stromal cells (BM-MSCs), under both physiological and pathological conditions. We demonstrate that PDGF-AA secreted by tumor cells induces CXCL5 expression in BM-MSCs by suppressing MDA-9-dependent YAP/MST signaling. CXCL5-derived tumor cell proliferation and immune suppression are consequences of the MDA-9/CXCL5 signaling axis, promoting PC disease progression. mda-9 knockout tumor cells express less PDGF-AA and do not develop bone metastases. Our data document a previously undefined role of MDA-9/Syntenin in the tumor and microenvironment in regulating PC bone metastasis. This study provides a framework for translational strategies to ameliorate health complications and morbidity associated with advanced PC.


Subject(s)
Bone Neoplasms , Melanoma , Prostatic Neoplasms , Male , Humans , Syntenins/genetics , Syntenins/metabolism , Melanoma/metabolism , Prostatic Neoplasms/genetics , Signal Transduction/genetics , Bone Neoplasms/genetics , Cell Line, Tumor , Tumor Microenvironment , Neoplasm Metastasis
2.
Proc Natl Acad Sci U S A ; 119(47): e2213835119, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36395141

ABSTRACT

Somatic copy number alterations (SCNAs), generally (1) losses containing interferons and interferon-pathway genes, many on chromosome 9p, predict immune-cold, immune checkpoint therapy (ICT)-resistant tumors (2); however, genomic regions mediating these effects are unclear and probably tissue specific. Previously, 9p21.3 loss was found to be an early genetic driver of human papillomavirus-negative (HPV-) head and neck squamous cancer (HNSC), associated with an immune-cold tumor microenvironment (TME) signal, and recent evidence suggested that this TME-cold phenotype was greatly enhanced with 9p21 deletion size, notably encompassing band 9p24.1 (3). Here, we report multi-omic, -threshold and continuous-variable dissection of 9p21 and 9p24 loci (including depth and degree of somatic alteration of each band at each locus, and each gene at each band) and TME of four HPV- HNSC cohorts. Preferential 9p24 deletion, CD8 T-cell immune-cold associations were observed, driven by 9p24.1 loss, and in turn by an essential telomeric regulatory gene element, JAK2-CD274. Surprisingly, same genetic region gains were immune hot. Related 9p21-TME analyses were less evident. Inherent 9p-band-level influences on anti-PD1 ICT survival rates, coincident with TME patterns, were also observed. At a 9p24.1 whole-transcriptome expression threshold of 60th percentile, ICT survival rate exceeded that of lower expression percentiles and of chemotherapy; below this transcript threshold, ICT survival was inferior to chemotherapy, the latter unaffected by 9p24.1 expression level (P-values < 0.01, including in a PD-L1 immunohistochemistry-positive patient subgroup). Whole-exome analyses of 10 solid-tumor types suggest that these 9p-related ICT findings could be relevant to squamous cancers, in which 9p24.1 gain/immune-hot associations exist.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Papillomavirus Infections , Humans , Tumor Microenvironment/genetics , Immune Checkpoint Inhibitors , Papillomavirus Infections/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics
3.
Genes Dev ; 31(12): 1212-1227, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28724615

ABSTRACT

In glioblastoma (GBM), heterogeneous expression of amplified and mutated epidermal growth factor receptor (EGFR) presents a substantial challenge for the effective use of EGFR-directed therapeutics. Here we demonstrate that heterogeneous expression of the wild-type receptor and its constitutively active mutant form, EGFRvIII, limits sensitivity to these therapies through an interclonal communication mechanism mediated by interleukin-6 (IL-6) cytokine secreted from EGFRvIII-positive tumor cells. IL-6 activates a NF-κB signaling axis in a paracrine and autocrine manner, leading to bromodomain protein 4 (BRD4)-dependent expression of the prosurvival protein survivin (BIRC5) and attenuation of sensitivity to EGFR tyrosine kinase inhibitors (TKIs). NF-κB and survivin are coordinately up-regulated in GBM patient tumors, and functional inhibition of either protein or BRD4 in in vitro and in vivo models restores sensitivity to EGFR TKIs. These results provide a rationale for improving anti-EGFR therapeutic efficacy through pharmacological uncoupling of a convergence point of NF-κB-mediated survival that is leveraged by an interclonal circuitry mechanism established by intratumoral mutational heterogeneity.


Subject(s)
Drug Resistance, Neoplasm/genetics , Glioblastoma/physiopathology , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction/genetics , Animals , Cell Communication , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Interleukin-6/metabolism , Mice , Mice, Nude , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
4.
J Cell Physiol ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38775127

ABSTRACT

Primary, glioblastoma, and secondary brain tumors, from metastases outside the brain, are among the most aggressive and therapeutically resistant cancers. A physiological barrier protecting the brain, the blood-brain barrier (BBB), functions as a deterrent to effective therapies. To enhance cancer therapy, we developed a cancer terminator virus (CTV), a unique tropism-modified adenovirus consisting of serotype 3 fiber knob on an otherwise Ad5 capsid that replicates in a cancer-selective manner and simultaneously produces a potent therapeutic cytokine, melanoma differentiation-associated gene-7/interleukin-24 (MDA-7/IL-24). A limitation of the CTV and most other viruses, including adenoviruses, is an inability to deliver systemically to treat brain tumors because of the BBB, nonspecific virus trapping, and immune clearance. These obstacles to effective viral therapy of brain cancer have now been overcome using focused ultrasound with a dual microbubble treatment, the focused ultrasound-double microbubble (FUS-DMB) approach. Proof-of-principle is now provided indicating that the BBB can be safely and transiently opened, and the CTV can then be administered in a second set of complement-treated microbubbles and released in the brain using focused ultrasound. Moreover, the FUS-DMB can be used to deliver the CTV multiple times in animals with glioblastoma  growing in their brain thereby resulting in a further enhancement in survival. This strategy permits efficient therapy of primary and secondary brain tumors enhancing animal survival without promoting harmful toxic or behavioral side effects. Additionally, when combined with a standard of care therapy, Temozolomide, a further increase in survival is achieved. The FUS-DMB approach with the CTV highlights a noninvasive strategy to treat brain cancers without surgery. This innovative delivery scheme combined with the therapeutic efficacy of the CTV provides a novel potential translational therapeutic approach for brain cancers.

5.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34016751

ABSTRACT

Melanoma differentiation associated gene-9 (MDA-9), Syntenin-1, or syndecan binding protein is a differentially regulated prometastatic gene with elevated expression in advanced stages of melanoma. MDA-9/Syntenin expression positively associates with advanced disease stage in multiple histologically distinct cancers and negatively correlates with patient survival and response to chemotherapy. MDA-9/Syntenin is a highly conserved PDZ-domain scaffold protein, robustly expressed in a spectrum of diverse cancer cell lines and clinical samples. PDZ domains interact with a number of proteins, many of which are critical regulators of signaling cascades in cancer. Knockdown of MDA-9/Syntenin decreases cancer cell metastasis, sensitizing these cells to radiation. Genetic silencing of MDA-9/Syntenin or treatment with a pharmacological inhibitor of the PDZ1 domain, PDZ1i, also activates the immune system to kill cancer cells. Additionally, suppression of MDA-9/Syntenin deregulates myeloid-derived suppressor cell differentiation via the STAT3/interleukin (IL)-1ß pathway, which concomitantly promotes activation of cytotoxic T lymphocytes. Biologically, PDZ1i treatment decreases metastatic nodule formation in the lungs, resulting in significantly fewer invasive cancer cells. In summary, our observations indicate that MDA-9/Syntenin provides a direct therapeutic target for mitigating aggressive breast cancer and a small-molecule inhibitor, PDZ1i, provides a promising reagent for inhibiting advanced breast cancer pathogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Interleukin-1beta/genetics , Lung Neoplasms/drug therapy , Oxadiazoles/pharmacology , Pyrimidines/pharmacology , Syntenins/genetics , Animals , Antineoplastic Agents/chemical synthesis , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Chemokine CCL11/genetics , Chemokine CCL11/immunology , Chemokine CCL17/genetics , Chemokine CCL17/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/immunology , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Oxadiazoles/chemical synthesis , Pyrimidines/chemical synthesis , Signal Transduction , Syntenins/antagonists & inhibitors , Syntenins/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Nature ; 543(7643): 122-125, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28178237

ABSTRACT

Human cells have twenty-three pairs of chromosomes. In cancer, however, genes can be amplified in chromosomes or in circular extrachromosomal DNA (ecDNA), although the frequency and functional importance of ecDNA are not understood. We performed whole-genome sequencing, structural modelling and cytogenetic analyses of 17 different cancer types, including analysis of the structure and function of chromosomes during metaphase of 2,572 dividing cells, and developed a software package called ECdetect to conduct unbiased, integrated ecDNA detection and analysis. Here we show that ecDNA was found in nearly half of human cancers; its frequency varied by tumour type, but it was almost never found in normal cells. Driver oncogenes were amplified most commonly in ecDNA, thereby increasing transcript level. Mathematical modelling predicted that ecDNA amplification would increase oncogene copy number and intratumoural heterogeneity more effectively than chromosomal amplification. We validated these predictions by quantitative analyses of cancer samples. The results presented here suggest that ecDNA contributes to accelerated evolution in cancer.


Subject(s)
DNA Copy Number Variations/genetics , Evolution, Molecular , Gene Amplification/genetics , Genetic Heterogeneity , Models, Genetic , Neoplasms/genetics , Oncogenes/genetics , Chromosomes, Human/genetics , Cytogenetic Analysis , DNA Mutational Analysis , Genome, Human/genetics , Humans , Metaphase/genetics , Neoplasms/classification , RNA, Messenger/analysis , RNA, Neoplasm/genetics , Reproducibility of Results , Software
7.
Mol Cell ; 60(2): 307-18, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26455392

ABSTRACT

Epidermal growth factor receptor (EGFR) gene amplification and mutations are the most common oncogenic events in glioblastoma (GBM), but the mechanisms by which they promote aggressive tumor growth are not well understood. Here, through integrated epigenome and transcriptome analyses of cell lines, genotyped clinical samples, and TCGA data, we show that EGFR mutations remodel the activated enhancer landscape of GBM, promoting tumorigenesis through a SOX9 and FOXG1-dependent transcriptional regulatory network in vitro and in vivo. The most common EGFR mutation, EGFRvIII, sensitizes GBM cells to the BET-bromodomain inhibitor JQ1 in a SOX9, FOXG1-dependent manner. These results identify the role of transcriptional/epigenetic remodeling in EGFR-dependent pathogenesis and suggest a mechanistic basis for epigenetic therapy.


Subject(s)
Brain Neoplasms/genetics , Epigenesis, Genetic , ErbB Receptors/genetics , Forkhead Transcription Factors/genetics , Glioblastoma/genetics , Nerve Tissue Proteins/genetics , SOX9 Transcription Factor/genetics , Adult , Animals , Azepines/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Child , ErbB Receptors/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Transcriptome , Triazoles/pharmacology
8.
Proc Natl Acad Sci U S A ; 117(22): 12324-12331, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32409605

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive cancer without currently effective therapies. Radiation and temozolomide (radio/TMZ) resistance are major contributors to cancer recurrence and failed GBM therapy. Heat shock proteins (HSPs), through regulation of extracellular matrix (ECM) remodeling and epithelial mesenchymal transition (EMT), provide mechanistic pathways contributing to the development of GBM and radio/TMZ-resistant GBM. The Friend leukemia integration 1 (Fli-1) signaling network has been implicated in oncogenesis in GBM, making it an appealing target for advancing novel therapeutics. Fli-1 is linked to oncogenic transformation with up-regulation in radio/TMZ-resistant GBM, transcriptionally regulating HSPB1. This link led us to search for targeted molecules that inhibit Fli-1. Expression screening for Fli-1 inhibitors identified lumefantrine, an antimalarial drug, as a probable Fli-1 inhibitor. Docking and isothermal calorimetry titration confirmed interaction between lumefantrine and Fli-1. Lumefantrine promoted growth suppression and apoptosis in vitro in parental and radio/TMZ-resistant GBM and inhibited tumor growth without toxicity in vivo in U87MG GBM and radio/TMZ-resistant GBM orthotopic tumor models. These data reveal that lumefantrine, an FDA-approved drug, represents a potential GBM therapeutic that functions through inhibition of the Fli-1/HSPB1/EMT/ECM remodeling protein networks.


Subject(s)
Antimalarials/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Lumefantrine/administration & dosage , Temozolomide/administration & dosage , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/radiotherapy , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
9.
Cancer Sci ; 113(5): 1555-1563, 2022 May.
Article in English | MEDLINE | ID: mdl-35271755

ABSTRACT

Cancer cells depend on metabolic reprogramming for survival, undergoing profound shifts in nutrient sensing, nutrient uptake and flux through anabolic pathways, in order to drive nucleotide, lipid, and protein synthesis and provide key intermediates needed for those pathways. Although metabolic enzymes themselves can be mutated, including to generate oncometabolites, this is a relatively rare event in cancer. Usually, gene amplification, overexpression, and/or downstream signal transduction upregulate rate-limiting metabolic enzymes and limit feedback loops, to drive persistent tumor growth. Recent molecular-genetic advances have revealed discrete links between oncogenotypes and the resultant metabolic phenotypes. However, more comprehensive approaches are needed to unravel the dynamic spatio-temporal regulatory map of enzymes and metabolites that enable cancer cells to adapt to their microenvironment to maximize tumor growth. Proteomic and metabolomic analyses are powerful tools for analyzing a repertoire of metabolic enzymes as well as intermediary metabolites, and in conjunction with other omics approaches could provide critical information in this regard. Here, we provide an overview of cancer metabolism, especially from an omics perspective and with a particular focus on the genomically well characterized malignant brain tumor, glioblastoma. We further discuss how metabolomics could be leveraged to improve the management of patients, by linking cancer cell genotype, epigenotype, and phenotype through metabolic reprogramming.


Subject(s)
Brain Neoplasms , Glioma , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Glioma/genetics , Glioma/metabolism , Humans , Metabolomics , Proteomics , Tumor Microenvironment
10.
Proc Natl Acad Sci U S A ; 116(12): 5687-5692, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30842276

ABSTRACT

Melanoma differentiation-associated gene-7/interleukin-24 (mda-7/IL-24) is a multifunctional cytokine displaying broad-spectrum anticancer activity in vitro or in vivo in preclinical animal cancer models and in a phase 1/2 clinical trial in patients with advanced cancers. mda-7/IL-24 targets specific miRNAs, including miR-221 and miR-320, for down-regulation in a cancer-selective manner. We demonstrate that mda-7/IL-24, administered through a replication incompetent type 5 adenovirus (Ad.mda-7) or with His-MDA-7/IL-24 protein, down-regulates DICER, a critical regulator in miRNA processing. This effect is specific for mature miR-221, as it does not affect Pri-miR-221 expression, and the DICER protein, as no changes occur in other miRNA processing cofactors, including DROSHA, PASHA, or Argonaute. DICER is unchanged by Ad.mda-7/IL-24 in normal immortal prostate cells, whereas Ad.mda-7 down-regulates DICER in multiple cancer cells including glioblastoma multiforme and prostate, breast, lung, and liver carcinoma cells. MDA-7/IL-24 protein down-regulates DICER expression through canonical IL-20/IL-22 receptors. Gain- and loss-of-function studies confirm that overexpression of DICER rescues deregulation of miRNAs by mda-7/IL-24, partially rescuing cancer cells from mda-7/IL-24-mediated cell death. Stable overexpression of DICER in cancer cells impedes Ad.mda-7 or His-MDA-7/IL-24 inhibition of cell growth, colony formation, PARP cleavage, and apoptosis. In addition, stable overexpression of DICER renders cancer cells more resistant to Ad.mda-7 inhibition of primary and secondary tumor growth. MDA-7/IL-24-mediated regulation of DICER is reactive oxygen species-dependent and mediated by melanogenesis-associated transcription factor. Our research uncovers a distinct role of mda-7/IL-24 in the regulation of miRNA biogenesis through alteration of the MITF-DICER pathway.


Subject(s)
DEAD-box RNA Helicases/metabolism , Interleukins/metabolism , MicroRNAs/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Ribonuclease III/metabolism , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , DEAD-box RNA Helicases/biosynthesis , DEAD-box RNA Helicases/genetics , Down-Regulation , Genes, Tumor Suppressor , Humans , Interleukins/genetics , Male , Mice , Mice, Nude , MicroRNAs/biosynthesis , Microphthalmia-Associated Transcription Factor/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/parasitology , Reactive Oxygen Species/metabolism , Ribonuclease III/biosynthesis , Ribonuclease III/genetics , Signal Transduction , Xenograft Model Antitumor Assays/methods
11.
Proc Natl Acad Sci U S A ; 116(20): 10009-10018, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31028138

ABSTRACT

Epidermal growth factor receptor (EGFR) signaling is initiated by a large ligand-favored conformational change of the extracellular domain (ECD) from a closed, self-inhibited tethered monomer, to an open untethered state, which exposes a loop required for strong dimerization and activation. In glioblastomas (GBMs), structurally heterogeneous missense and deletion mutations concentrate at the ECD for unclear reasons. We explore the conformational impact of GBM missense mutations, combining elastic network models (ENMs) with multiple molecular dynamics (MD) trajectories. Our simulations reveal that the main missense class, located at the I-II interface away from the self-inhibitory tether, can unexpectedly favor spontaneous untethering to a compact intermediate state, here validated by small-angle X-ray scattering (SAXS). Significantly, such intermediate is characterized by the rotation of a large ECD fragment (N-TR1), deleted in the most common GBM mutation, EGFRvIII, and that makes accessible a cryptic epitope characteristic of cancer cells. This observation suggested potential structural equivalence of missense and deletion ECD changes in GBMs. Corroborating this hypothesis, our FACS, in vitro, and in vivo data demonstrate that entirely different ECD variants all converge to remove N-TR1 steric hindrance from the 806-epitope, which we show is allosterically coupled to an intermediate kinase and hallmarks increased oncogenicity. Finally, the detected extraintracellular coupling allows for synergistic cotargeting of the intermediate with mAb806 and inhibitors, which is proved herein.


Subject(s)
Genes, erbB-1 , Glioblastoma/genetics , Epitopes , HEK293 Cells , Humans , Mutation, Missense
12.
Proc Natl Acad Sci U S A ; 116(13): 6308-6312, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30858324

ABSTRACT

Liquid biopsies, based on cell free DNA (cfDNA) and proteins, have shown the potential to detect early stage cancers of diverse tissue types. However, most of these studies were retrospective, using individuals previously diagnosed with cancer as cases and healthy individuals as controls. Here, we developed a liquid biopsy assay, named the hepatocellular carcinoma screen (HCCscreen), to identify HCC from the surface antigen of hepatitis B virus (HBsAg) positive asymptomatic individuals in the community population. The training cohort consisted of individuals who had liver nodules and/or elevated serum α-fetoprotein (AFP) levels, and the assay robustly separated those with HCC from those who were non-HCC with a sensitivity of 85% and a specificity of 93%. We further applied this assay to 331 individuals with normal liver ultrasonography and serum AFP levels. A total of 24 positive cases were identified, and a clinical follow-up for 6-8 mo confirmed four had developed HCC. No HCC cases were diagnosed from the 307 test-negative individuals in the follow-up during the same timescale. Thus, the assay showed 100% sensitivity, 94% specificity, and 17% positive predictive value in the validation cohort. Notably, each of the four HCC cases was at the early stage (<3 cm) when diagnosed. Our study provides evidence that the use of combined detection of cfDNA alterations and protein markers is a feasible approach to identify early stage HCC from asymptomatic community populations with unknown HCC status.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/diagnosis , Early Detection of Cancer/methods , Hepatitis B Surface Antigens/blood , Liquid Biopsy/methods , Liver Neoplasms/diagnosis , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Cell-Free Nucleic Acids , Hepatitis B virus , Hepatitis B, Chronic , Humans , Liver Neoplasms/blood , Liver Neoplasms/pathology , Sensitivity and Specificity , Ultrasonography
13.
Proc Natl Acad Sci U S A ; 115(22): 5768-5773, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29760085

ABSTRACT

Glioma stem cells (GSCs) comprise a small subpopulation of glioblastoma multiforme cells that contribute to therapy resistance, poor prognosis, and tumor recurrence. Protective autophagy promotes resistance of GSCs to anoikis, a form of programmed cell death occurring when anchorage-dependent cells detach from the extracellular matrix. In nonadherent conditions, GSCs display protective autophagy and anoikis-resistance, which correlates with expression of melanoma differentiation associated gene-9/Syntenin (MDA-9) (syndecan binding protein; SDCBP). When MDA-9 is suppressed, GSCs undergo autophagic death supporting the hypothesis that MDA-9 regulates protective autophagy in GSCs under anoikis conditions. MDA-9 maintains protective autophagy through phosphorylation of BCL2 and by suppressing high levels of autophagy through EGFR signaling. MDA-9 promotes these changes by modifying FAK and PKC signaling. Gain-of-function and loss-of-function genetic approaches demonstrate that MDA-9 regulates pEGFR and pBCL2 expression through FAK and pPKC. EGFR signaling inhibits autophagy markers (ATG5, Lamp1, LC3B), helping to maintain protective autophagy, and along with pBCL2 maintain survival of GSCs. In the absence of MDA-9, this protective mechanism is deregulated; EGFR no longer maintains protective autophagy, leading to highly elevated and sustained levels of autophagy and consequently decreased cell survival. In addition, pBCL2 is down-regulated in the absence of MDA-9, leading to cell death in GSCs under conditions of anoikis. Our studies confirm a functional link between MDA-9 expression and protective autophagy in GSCs and show that inhibition of MDA-9 reverses protective autophagy and induces anoikis and cell death in GSCs.


Subject(s)
Anoikis/genetics , Autophagy/genetics , Drug Resistance, Neoplasm/genetics , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Syntenins/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioma/genetics , Humans , Syntenins/genetics , Tumor Cells, Cultured
14.
Int J Mol Sci ; 23(1)2021 Dec 22.
Article in English | MEDLINE | ID: mdl-35008495

ABSTRACT

Melanoma differentiation associated gene-7/interleukin-24 (MDA-7/IL-24), a secreted protein of the IL-10 family, was first identified more than two decades ago as a novel gene differentially expressed in terminally differentiating human metastatic melanoma cells. MDA-7/IL-24 functions as a potent tumor suppressor exerting a diverse array of functions including the inhibition of tumor growth, invasion, angiogenesis, and metastasis, and induction of potent "bystander" antitumor activity and synergy with conventional cancer therapeutics. MDA-7/IL-24 induces cancer-specific cell death through apoptosis or toxic autophagy, which was initially established in vitro and in preclinical animal models in vivo and later in a Phase I clinical trial in patients with advanced cancers. This review summarizes the history and our current understanding of the molecular/biological mechanisms of MDA-7/IL-24 action rendering it a potent cancer suppressor.


Subject(s)
Interleukins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/physiology , Autophagy/physiology , Cell Death/physiology , Humans , Melanoma/metabolism
15.
J Biol Chem ; 294(51): 19740-19751, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31712311

ABSTRACT

In cancer, aberrant growth factor receptor signaling reprograms cellular metabolism and global gene transcription to drive aggressive growth, but the underlying mechanisms are not well-understood. Here we show that in the highly lethal brain tumor glioblastoma (GBM), mTOR complex 2 (mTORC2), a critical core component of the growth factor signaling system, couples acetyl-CoA production with nuclear translocation of histone-modifying enzymes including pyruvate dehydrogenase and class IIa histone deacetylases to globally alter histone acetylation. Integrated analyses in orthotopic mouse models and in clinical GBM samples reveal that mTORC2 controls iron metabolisms via histone H3 acetylation of the iron-related gene promoter, promoting tumor cell survival. These results nominate mTORC2 as a critical epigenetic regulator of iron metabolism in cancer.


Subject(s)
Brain Neoplasms/metabolism , Epigenesis, Genetic , Glioblastoma/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Iron/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , Histones/chemistry , Humans , Immediate-Early Proteins/metabolism , Metabolome , Mice , Neoplasm Transplantation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Signal Transduction
16.
Pharmacol Res ; 155: 104695, 2020 05.
Article in English | MEDLINE | ID: mdl-32061839

ABSTRACT

The primary cause of cancer-related death from solid tumors is metastasis. While unraveling the mechanisms of this complicated process continues, our ability to effectively target and treat it to decrease patient morbidity and mortality remains disappointing. Early detection of metastatic lesions and approaches to treat metastases (both pharmacological and genetic) are of prime importance to obstruct this process clinically. Metastasis is complex involving both genetic and epigenetic changes in the constantly evolving tumor cell. Moreover, many discrete steps have been identified in metastatic spread, including invasion, intravasation, angiogenesis, attachment at a distant site (secondary seeding), extravasation and micrometastasis and tumor dormancy development. Here, we provide an overview of the metastatic process and highlight a unique pro-metastatic gene, melanoma differentiation associated gene-9/Syntenin (MDA-9/Syntenin) also called syndecan binding protein (SDCBP), which is a major contributor to the majority of independent metastatic events. MDA-9 expression is elevated in a wide range of carcinomas and other cancers, including melanoma, glioblastoma multiforme and neuroblastoma, suggesting that it may provide an appropriate target to intervene in metastasis. Pre-clinical studies confirm that inhibiting MDA-9 either genetically or pharmacologically profoundly suppresses metastasis. An additional benefit to blocking MDA-9 in metastatic cells is sensitization of these cells to a second therapeutic agent, which converts anti-invasion effects to tumor cytocidal effects. Continued mechanistic and therapeutic insights hold promise to advance development of truly effective therapies for metastasis in the future.


Subject(s)
Neoplasm Metastasis/genetics , Neoplasms/therapy , Syntenins/genetics , Animals , Humans , Neoplasms/genetics , Neoplasms/pathology
17.
Proc Natl Acad Sci U S A ; 114(26): 6812-6817, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28607091

ABSTRACT

Oncolytic virotherapy is a treatment modality that uses native or genetically modified viruses that selectively replicate in and kill tumor cells. Viruses represent a type of pathogen-associated molecular pattern and thereby induce the up-regulation of dozens of cytokines via activating the host innate immune system. Second mitochondria-derived activator of caspases (Smac) mimetic compounds (SMCs), which antagonize the function of inhibitor of apoptosis proteins (IAPs) and induce apoptosis, sensitize tumor cells to multiple cytokines. Therefore, we sought to determine whether SMCs sensitize tumor cells to cytokines induced by the oncolytic M1 virus, thus enhancing a bystander killing effect. Here, we report that SMCs potentiate the oncolytic effect of M1 in vitro, in vivo, and ex vivo. This strengthened oncolytic efficacy resulted from the enhanced bystander killing effect caused by the M1 virus via cytokine induction. Through a microarray analysis and subsequent validation using recombinant cytokines, we identified IL-8, IL-1A, and TRAIL as the key cytokines in the bystander killing effect. Furthermore, SMCs increased the replication of M1, and the accumulation of virus protein induced irreversible endoplasmic reticulum stress- and c-Jun N-terminal kinase-mediated apoptosis. Nevertheless, the combined treatment with M1 and SMCs had little effect on normal and human primary cells. Because SMCs selectively and significantly enhance the bystander killing effect and the replication of oncolytic virus M1 specifically in cancer cells, this combined treatment may represent a promising therapeutic strategy.


Subject(s)
Apoptosis/drug effects , Bystander Effect/drug effects , Neoplasms, Experimental/therapy , Oligopeptides/pharmacology , Oncolytic Viruses/physiology , Peptidomimetics/pharmacology , Virus Replication/drug effects , Animals , Apoptosis/immunology , Bystander Effect/immunology , Cell Line, Tumor , Cytokines/immunology , Humans , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
18.
Proc Natl Acad Sci U S A ; 114(2): 370-375, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28011764

ABSTRACT

Glioblastoma multiforme (GBM) is an intractable tumor despite therapeutic advances, principally because of its invasive properties. Radiation is a staple in therapeutic regimens, although cells surviving radiation can become more aggressive and invasive. Subtraction hybridization identified melanoma differentiation-associated gene 9 [MDA-9/Syntenin; syndecan-binding protein (SDCBP)] as a differentially regulated gene associated with aggressive cancer phenotypes in melanoma. MDA-9/Syntenin, a highly conserved double-PDZ domain-containing scaffolding protein, is robustly expressed in human-derived GBM cell lines and patient samples, with expression increasing with tumor grade and correlating with shorter survival times and poorer response to radiotherapy. Knockdown of MDA-9/Syntenin sensitizes GBM cells to radiation, reducing postradiation invasion gains. Radiation induces Src and EGFRvIII signaling, which is abrogated through MDA-9/Syntenin down-regulation. A specific inhibitor of MDA-9/Syntenin activity, PDZ1i (113B7), identified through NMR-guided fragment-based drug design, inhibited MDA-9/Syntenin binding to EGFRvIII, which increased following radiation. Both genetic (shmda-9) and pharmacological (PDZ1i) targeting of MDA-9/Syntenin reduced invasion gains in GBM cells following radiation. Although not affecting normal astrocyte survival when combined with radiation, PDZ1i radiosensitized GBM cells. PDZ1i inhibited crucial GBM signaling involving FAK and mutant EGFR, EGFRvIII, and abrogated gains in secreted proteases, MMP-2 and MMP-9, following radiation. In an in vivo glioma model, PDZ1i resulted in smaller, less invasive tumors and enhanced survival. When combined with radiation, survival gains exceeded radiotherapy alone. MDA-9/Syntenin (SDCBP) provides a direct target for therapy of aggressive cancers such as GBM, and defined small-molecule inhibitors such as PDZ1i hold promise to advance targeted brain cancer therapy.


Subject(s)
Glioblastoma/genetics , Neoplasm Invasiveness/genetics , Syntenins/genetics , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Melanoma/genetics , Mice , Mice, Nude , PDZ Domains/genetics , Signal Transduction/genetics , src-Family Kinases/genetics
19.
Genes Dev ; 26(8): 756-84, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22508724

ABSTRACT

Glioblastoma is both the most common and lethal primary malignant brain tumor. Extensive multiplatform genomic characterization has provided a higher-resolution picture of the molecular alterations underlying this disease. These studies provide the emerging view that "glioblastoma" represents several histologically similar yet molecularly heterogeneous diseases, which influences taxonomic classification systems, prognosis, and therapeutic decisions.


Subject(s)
Brain Neoplasms/classification , Brain Neoplasms/genetics , Glioblastoma/classification , Glioblastoma/genetics , Brain Neoplasms/pathology , Gene Expression Profiling , Genes, Tumor Suppressor , Genomics , Glioblastoma/pathology , Humans , Neovascularization, Pathologic/genetics , Transcription, Genetic
20.
Neuropathology ; 39(1): 3-13, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30609184

ABSTRACT

Cancer is a genetic disease that is currently classified not only by its tissue and cell type of origin but increasingly by its molecular composition. Increasingly, tumor classification and subtyping is being performed based upon the oncogene gains, tumor suppressor losses, and associated epigenetic and transcriptional features. However, cancers, including brain tumors, are also characterized by profound alterations in cellular metabolism. At present, even though signature mutations in known metabolic enzymes are recognized as being important, the metabolic landscape of tumors is not currently incorporated into tumor diagnostic categories. Here we describe a set of recent discoveries on metabolic reprogramming driven by mutations in the genes for the isocitrate dehydrogenase (IDH) and receptor tyrosine kinase (RTK) pathways, which are the most commonly observed aberrations in diffuse gliomas. We highlight the importance of oncometabolites to dynamically shift the epigenetic landscape in IDH-mutant gliomas, and c-Myc and mechanistic target of rapamycin (mTOR) complexes in RTK-mutated gliomas to adapt to the microenvironment through metabolic reprogramming. These signify the integration of the genetic mutations with metabolic reprogramming and epigenetic shifts in diffuse gliomas, shedding new light onto potential patient subsets, coupled with information to guide the development of new therapeutic opportunities against the deadly types of brain tumors.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Animals , Brain Neoplasms/genetics , Epigenesis, Genetic , Glioma/genetics , Humans , Isocitrate Dehydrogenase/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL