Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Cell ; 183(2): 377-394.e21, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32976798

ABSTRACT

We employed scRNA sequencing to extensively characterize the cellular landscape of human liver from development to disease. Analysis of ∼212,000 cells representing human fetal, hepatocellular carcinoma (HCC), and mouse liver revealed remarkable fetal-like reprogramming of the tumor microenvironment. Specifically, the HCC ecosystem displayed features reminiscent of fetal development, including re-emergence of fetal-associated endothelial cells (PLVAP/VEGFR2) and fetal-like (FOLR2) tumor-associated macrophages. In a cross-species comparative analysis, we discovered remarkable similarity between mouse embryonic, fetal-liver, and tumor macrophages. Spatial transcriptomics further revealed a shared onco-fetal ecosystem between fetal liver and HCC. Furthermore, gene regulatory analysis, spatial transcriptomics, and in vitro functional assays implicated VEGF and NOTCH signaling in maintaining onco-fetal ecosystem. Taken together, we report a shared immunosuppressive onco-fetal ecosystem in fetal liver and HCC. Our results unravel a previously unexplored onco-fetal reprogramming of the tumor ecosystem, provide novel targets for therapeutic interventions in HCC, and open avenues for identifying similar paradigms in other cancers and disease.


Subject(s)
Carcinoma, Hepatocellular/pathology , Endothelial Cells/metabolism , Tumor Microenvironment/genetics , Adult , Animals , Carcinoma, Hepatocellular/genetics , Cell Line , Disease Models, Animal , Endothelial Cells/pathology , Female , Folate Receptor 2/metabolism , Gene Expression Profiling/methods , Humans , Liver/pathology , Liver Neoplasms/genetics , Macrophages/metabolism , Male , Membrane Proteins/metabolism , Mice , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/genetics , Transcriptome/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Nucleic Acids Res ; 51(1): 1-16, 2023 01 11.
Article in English | MEDLINE | ID: mdl-35697349

ABSTRACT

Transcriptional reactivation of hTERT is the limiting step in tumorigenesis. While mutations in hTERT promoter present in 19% of cancers are recognized as key drivers of hTERT reactivation, mechanisms by which wildtype hTERT (WT-hTERT) promoter is reactivated, in majority of human cancers, remain unknown. Using primary colorectal cancers (CRC) we identified Tert INTeracting region 2 (T-INT2), the critical chromatin region essential for reactivating WT-hTERT promoter in CRCs. Elevated ß-catenin and JunD level in CRC facilitates chromatin interaction between hTERT promoter and T-INT2 that is necessary to turn on hTERTexpression. Pharmacological screens uncovered salinomycin, which inhibits JunD mediated hTERT-T-INT2 interaction that is required for the formation of a stable transcription complex on the hTERT promoter. Our results showed for the first time how known CRC alterations, such as APC, lead to WT-hTERT promoter reactivation during stepwise-tumorigenesis and provide a new perspective for developing cancer-specific drugs.


Healthy and cancer cells harbor the same DNA sequence, but reactivation of the Human Telomerase Reverse Transcriptase (hTERT) gene is observed only in cancer cells. How does that happen was not known for over three decades of research? This study identifies a specific DNA structure that forms only in cancer cells and brings the necessary molecular machinery into the correct position to activate the hTERT gene. The detailed mechanism of hTERT activation provided in this study will be instrumental in designing cancer cell-specific hTERT inhibitors, especially since all the other ways of inhibiting telomerase failed in the clinic.


Subject(s)
Colorectal Neoplasms , Telomerase , Humans , Carcinogenesis , Chromatin/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Promoter Regions, Genetic , Telomerase/antagonists & inhibitors , Telomerase/genetics , Transcription, Genetic
3.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35027447

ABSTRACT

Cancer-specific hTERT promoter mutations reported in 19% of cancers result in enhanced telomerase activity. Understanding the distinctions between transcriptional regulation of wild-type (WT) and mutant (Mut) hTERT promoters may open up avenues for development of inhibitors which specially block hTERT expression in cancer cells. To comprehensively identify physiological regulators of WT- or Mut-hTERT promoters, we generated several isogenic reporter cells driven by endogenous hTERT loci. Genome-wide CRISPR-Cas9 and small interfering RNA screens using these isogenic reporter lines identified specific regulators of Mut-hTERT promoters. We validate and characterize one of these hits, namely, MED12, a kinase subunit of mediator complex. We demonstrate that MED12 specifically drives expression of hTERT from the Mut-hTERT promoter by mediating long-range chromatin interaction between the proximal Mut-hTERT promoter and T-INT1 distal regulatory region 260 kb upstream. Several hits identified in our screens could serve as potential therapeutic targets, inhibition of which may specifically block Mut-hTERT promoter driven telomerase reactivation in cancers.


Subject(s)
Mutation , Promoter Regions, Genetic , Telomerase/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Chromatin , DNA-Binding Proteins , Gene Editing , Gene Expression Regulation, Neoplastic , Humans , Mediator Complex/genetics , Mediator Complex/metabolism , Neoplasms/genetics , Regulatory Sequences, Nucleic Acid , Telomerase/metabolism , Transcription Factors , Transcription, Genetic
4.
J Hepatol ; 81(1): 42-61, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38423478

ABSTRACT

BACKGROUND & AIMS: Hepatitis B surface antigen (HBsAg) loss or functional cure (FC) is considered the optimal therapeutic outcome for patients with chronic hepatitis B (CHB). However, the immune-pathological biomarkers and underlying mechanisms of FC remain unclear. In this study we comprehensively interrogate disease-associated cell states identified within intrahepatic tissue and matched PBMCs (peripheral blood mononuclear cells) from patients with CHB or after FC, at the resolution of single cells, to provide novel insights into putative mechanisms underlying FC. METHODS: We combined single-cell transcriptomics (single-cell RNA sequencing) with multiparametric flow cytometry-based immune phenotyping, and multiplexed immunofluorescence to elucidate the immunopathological cell states associated with CHB vs. FC. RESULTS: We found that the intrahepatic environment in CHB and FC displays specific cell identities and molecular signatures that are distinct from those found in matched PBMCs. FC is associated with the emergence of an altered adaptive immune response marked by CD4 cytotoxic T lymphocytes, and an activated innate response represented by liver-resident natural killer cells, specific Kupffer cell subtypes and marginated neutrophils. Surprisingly, we found MHC class II-expressing hepatocytes in patients achieving FC, as well as low but persistent levels of covalently closed circular DNA and pregenomic RNA, which may play an important role in FC. CONCLUSIONS: Our study provides conceptually novel insights into the immuno-pathological control of HBV cure, and opens exciting new avenues for clinical management, biomarker discovery and therapeutic development. We believe that the discoveries from this study, as it relates to the activation of an innate and altered immune response that may facilitate sustained, low-grade inflammation, may have broader implications in the resolution of chronic viral hepatitis. IMPACT AND IMPLICATIONS: This study dissects the immuno-pathological cell states associated with functionally cured chronic hepatitis B (defined by the loss of HBV surface antigen or HBsAg). We identified the sustained presence of very low viral load, accessory antigen-presenting hepatocytes, adaptive-memory-like natural killer cells, and the emergence of helper CD4 T cells with cytotoxic or effector-like signatures associated with functional cure, suggesting previously unsuspected alterations in the adaptive immune response, as well as a key role for the innate immune response in achieving or maintaining functional cure. Overall, the insights generated from this study may provide new avenues for the development of alternative therapies as well as patient surveillance for better clinical management of chronic hepatitis B.


Subject(s)
Adaptive Immunity , Hepatitis B, Chronic , Immunity, Innate , Single-Cell Analysis , Humans , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Immunity, Innate/immunology , Adaptive Immunity/immunology , Single-Cell Analysis/methods , Hepatitis B virus/immunology , Hepatitis B virus/genetics , Male , Female , T-Lymphocytes, Cytotoxic/immunology , Adult , Liver/immunology , Liver/pathology , Hepatitis B Surface Antigens/immunology , Middle Aged , Killer Cells, Natural/immunology
5.
Cell Mol Life Sci ; 78(6): 2607-2619, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33388834

ABSTRACT

Transfer RNA (tRNA)-derived fragments (tRFs) are an emerging class of conserved small non-coding RNAs that play important roles in post-transcriptional gene regulation. High-throughput sequencing of multiple biological samples have identified heterogeneous species of tRFs with distinct functionalities. These small RNAs have garnered a lot of scientific attention due to their ubiquitous expression and versatility in regulating various biological processes. In this review, we highlight our current understanding of tRF biogenesis and their regulatory functions. We summarize the diverse modes of biogenesis through which tRFs are generated and discuss the mechanism through which different tRF species regulate gene expression and the biological implications. Finally, we conceptualize research areas that require focus to strengthen our understanding of the biogenesis and function of tRFs.


Subject(s)
RNA, Small Untranslated/metabolism , RNA, Transfer/metabolism , Animals , Gene Expression Regulation , Humans , Neoplasms/genetics , Neoplasms/pathology , Plants/genetics , Plants/metabolism , RNA Processing, Post-Transcriptional , RNA, Small Untranslated/genetics , RNA, Transfer/genetics , Ribonucleoproteins/metabolism
6.
EMBO Rep ; 20(7): e47789, 2019 07.
Article in English | MEDLINE | ID: mdl-31267708

ABSTRACT

Transfer RNA (tRNA)-derived small RNAs (tsRNAs) have recently emerged as important regulators of protein translation and shown to have diverse biological functions. However, the underlying cellular and molecular mechanisms of tsRNA function in the context of dynamic cell-state transitions remain unclear. Expression analysis of tsRNAs in distinct heterologous cell and tissue models of stem vs. differentiated states revealed a differentiation-dependent enrichment of 5'-tsRNAs. We report the identification of a set of 5'-tsRNAs that is upregulated in differentiating mouse embryonic stem cells (mESCs). Notably, interactome studies with differentially enriched 5'-tsRNAs revealed a switch in their association with "effector" RNPs and "target" mRNAs in different cell states. We demonstrate that specific 5'-tsRNAs can preferentially interact with the RNA-binding protein, Igf2bp1, in the RA-induced differentiated state. This association influences the transcript stability and thereby translation of the pluripotency-promoting factor, c-Myc, thus providing a mechanistic basis for how 5'-tsRNAs can modulate stem cell states in mESCs. Together our study highlights the role of 5'-tsRNAs in defining distinct cell states.


Subject(s)
Cell Differentiation , MicroRNAs/metabolism , RNA, Transfer/metabolism , Animals , Cells, Cultured , HCT116 Cells , Humans , Mice , MicroRNAs/genetics , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Stability , RNA, Transfer/genetics , RNA-Binding Proteins/metabolism
7.
Anal Chem ; 91(2): 1277-1285, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30362713

ABSTRACT

In the context of tumor analysis, the implementation of precision medicine requires on-time clinical measurements, which requires rapid large-scale single-cell screening that obtains cell population distributions and functions in tumors to determine disease progression for therapeutics. In this study, a high-throughput screening (HTS) platform integrating optical fluorescence detectors and a computational method was developed as a droplet-based microfluidic flow cytometer (Droplet-µFC) to comprehensively analyze multiple proteolytic activities of a patient-derived tumor (with ∼0.5-2 M cells) at single-cell resolution within 2 h. The data-driven analytical method identified distinct cell types and status through protease profiling with high precision. Multiple protease activities of single cells harvested from a tumor were thus determined with a throughput of ∼100 cells per second. This platform was used to screen protease activities of a wide range of cell types, forming a library. With the development of advanced computational clustering and cell mapping, rapid quantitative tumor profiling with a comprehensive description of cell population distributions and functions could be obtained for clinical treatments.


Subject(s)
Flow Cytometry/methods , Microfluidic Analytical Techniques/methods , Neoplasms/enzymology , Peptide Hydrolases/analysis , Animals , Antineoplastic Agents , Cell Line, Tumor , Databases, Factual , Erlotinib Hydrochloride/pharmacology , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/metabolism , High-Throughput Screening Assays , Humans , Lab-On-A-Chip Devices , Mice , Neoplasms/pathology , Oligopeptides/metabolism , Peptide Hydrolases/metabolism , Proteolysis , Single-Cell Analysis/methods
8.
Stem Cells ; 35(8): 1924-1933, 2017 08.
Article in English | MEDLINE | ID: mdl-28577307

ABSTRACT

The 2i-media, composed of two small molecule inhibitors (PD0325901 and CHIR99021) against MEK and GSK3-kinases, respectively, is known to establish naïve ground state pluripotency in mouse embryonic stem cells (mESCs). These inhibitors block MEK-mediated differentiation, while driving ß-catenin dependent de-repression of pluripotency promoting targets. However, accumulating evidence suggest that ß-catenin's association with activating TCFs (TCF7 and TCF7L2) can induce expression of several lineage-specific prodifferentiation genes. We posited that CHIR-induced upregulation of ß-catenin levels could therefore compromise the stability of the naïve state in long-term cultures. Here, we investigated whether replacing CHIR with iCRT3, a small molecule that abrogates ß-catenin-TCF interaction, can still retain ground state pluripotency in mESCs. Our data suggests that iCRT3 + PD mediated coinhibition of MEK and ß-catenin/TCF-dependent transcriptional activity over multiple passages significantly reduces expression of differentiation markers, as compared to 2i. Furthermore, the ability to efficiently contribute toward chimera generation and germline transmission suggests that the inhibition of ß-catenin's TCF-dependent transcriptional activity, independent of its protein expression level, retains the naïve ground state pluripotency in mESCs. Additionally, growth medium containing iCRT3 + PD can provide an alternative to 2i as a stable culture method. Stem Cells 2017;35:1924-1933.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , beta Catenin/metabolism , Animals , Benzamides/pharmacology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Female , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Oxazoles/pharmacology , Pluripotent Stem Cells/drug effects , Protein Binding/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Transcriptome/drug effects , Transcriptome/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Nucleic Acids Res ; 44(D1): D969-74, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26496946

ABSTRACT

In order to advance our understanding of colorectal cancer (CRC) development and progression, biomedical researchers have generated large amounts of OMICS data from CRC patient samples and representative cell lines. However, these data are deposited in various repositories or in supplementary tables. A database which integrates data from heterogeneous resources and enables analysis of the multidimensional data sets, specifically pertaining to CRC is currently lacking. Here, we have developed Colorectal Cancer Atlas (http://www.colonatlas.org), an integrated web-based resource that catalogues the genomic and proteomic annotations identified in CRC tissues and cell lines. The data catalogued to-date include sequence variations as well as quantitative and non-quantitative protein expression data. The database enables the analysis of these data in the context of signaling pathways, protein-protein interactions, Gene Ontology terms, protein domains and post-translational modifications. Currently, Colorectal Cancer Atlas contains data for >13 711 CRC tissues, >165 CRC cell lines, 62 251 protein identifications, >8.3 million MS/MS spectra, >18 410 genes with sequence variations (404 278 entries) and 351 pathways with sequence variants. Overall, Colorectal Cancer Atlas has been designed to serve as a central resource to facilitate research in CRC.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Databases, Genetic , Genomics , Proteomics , Cell Line, Tumor , Humans , Molecular Sequence Annotation , Mutation , Protein Processing, Post-Translational , Protein Structure, Tertiary
10.
Development ; 140(6): 1171-83, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23444350

ABSTRACT

The maintenance of pluripotency in mouse embryonic stem cells (mESCs) relies on the activity of a transcriptional network that is fuelled by the activity of three transcription factors (Nanog, Oct4 and Sox2) and balanced by the repressive activity of Tcf3. Extracellular signals modulate the activity of the network and regulate the differentiation capacity of the cells. Wnt/ß-catenin signaling has emerged as a significant potentiator of pluripotency: increases in the levels of ß-catenin regulate the activity of Oct4 and Nanog, and enhance pluripotency. A recent report shows that ß-catenin achieves some of these effects by modulating the activity of Tcf3, and that this effect does not require its transcriptional activation domain. Here, we show that during self-renewal there is negligible transcriptional activity of ß-catenin and that this is due to its tight association with membranes, where we find it in a complex with Oct4 and E-cadherin. Differentiation triggers a burst of Wnt/ß-catenin transcriptional activity that coincides with the disassembly of the complex. Our results establish that ß-catenin, but not its transcriptional activity, is central to pluripotency acting through a ß-catenin/Oct4 complex.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/physiology , beta Catenin/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/ultrastructure , Gene Expression Regulation, Developmental/drug effects , Mice , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , Octamer Transcription Factor-3/physiology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , RNA, Small Interfering/pharmacology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/physiology
11.
Proc Natl Acad Sci U S A ; 110(39): 15710-5, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-24019458

ABSTRACT

Androgen receptor (AR) is the major therapeutic target in aggressive prostate cancer. However, targeting AR alone can result in drug resistance and disease recurrence. Therefore, simultaneous targeting of multiple pathways could in principle be an effective approach to treating prostate cancer. Here we provide proof-of-concept that a small-molecule inhibitor of nuclear ß-catenin activity (called C3) can inhibit both the AR and ß-catenin-signaling pathways that are often misregulated in prostate cancer. Treatment with C3 ablated prostate cancer cell growth by disruption of both ß-catenin/T-cell factor and ß-catenin/AR protein interaction, reflecting the fact that T-cell factor and AR have overlapping binding sites on ß-catenin. Given that AR interacts with, and is transcriptionally regulated by ß-catenin, C3 treatment also resulted in decreased occupancy of ß-catenin on the AR promoter and diminished AR and AR/ß-catenin target gene expression. Interestingly, C3 treatment resulted in decreased AR binding to target genes accompanied by decreased recruitment of an AR and ß-catenin cofactor, coactivator-associated arginine methyltransferase 1 (CARM1), providing insight into the unrecognized function of ß-catenin in prostate cancer. Importantly, C3 inhibited tumor growth in an in vivo xenograft model and blocked renewal of bicalutamide-resistant sphere-forming cells, indicating the therapeutic potential of this approach.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , beta Catenin/antagonists & inhibitors , Animals , Biomarkers, Tumor/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Binding/drug effects , Protein Binding/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays , beta Catenin/metabolism
12.
Am J Respir Crit Care Med ; 190(2): 185-95, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24921217

ABSTRACT

RATIONALE: Wnt/ß-catenin signaling has been implicated in lung fibrosis, but how this occurs and whether expression changes in Wnt pathway components predict disease progression is unknown. OBJECTIVES: To determine whether the Wnt coreceptor Lrp5 drives pulmonary fibrosis in mice and is predictive of disease severity in humans. METHODS: We examined mice with impaired Wnt signaling caused by loss of the Wnt coreceptor Lrp5 in models of lung fibrosis induced by bleomycin or an adenovirus encoding an active form of transforming growth factor (TGF)-ß. We also analyzed gene expression in peripheral blood mononuclear cells (PBMC) from patients with idiopathic pulmonary fibrosis (IPF). MEASUREMENTS AND MAIN RESULTS: In patients with IPF, analysis of peripheral blood mononuclear cells revealed that elevation of positive regulators, Lrp5 and 6, was independently associated with disease progression. LRP5 was also associated with disease severity at presentation in an additional cohort of patients with IPF. Lrp5 null mice were protected against bleomycin-induced pulmonary fibrosis, an effect that was phenocopied by direct inhibition of ß-catenin signaling by the small molecular inhibitor of ß-catenin responsive transcription. Transplantation of Lrp5 null bone marrow cells into wild-type mice did not limit fibrosis. Instead, Lrp5 loss was associated with reduced TGF-ß production by alveolar type 2 cells and leukocytes. Consistent with a role of Lrp5 in the activation of TGF-ß, Lrp5 null mice were not protected against lung fibrosis induced by TGF-ß. CONCLUSIONS: We show that the Wnt coreceptor, Lrp5, is a genetic driver of lung fibrosis in mice and a marker of disease progression and severity in humans with IPF. Evidence that TGF-ß signaling can override a loss in Lrp5 has implications for patient selection and timing of Wnt pathway inhibitors in lung fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Aged , Animals , Biomarkers/metabolism , Disease Progression , Female , Humans , Idiopathic Pulmonary Fibrosis/etiology , Leukocytes, Mononuclear/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Prospective Studies , Severity of Illness Index , Signal Transduction , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
13.
Br J Haematol ; 167(1): 87-99, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24995804

ABSTRACT

While childhood acute lymphoblastic leukaemia (ALL) is now highly curable, the dismal prognosis for children who relapse warrants novel therapeutic approaches. Previously, using an integrated genomic analysis of matched diagnosis-relapse paired samples, we identified overactivation of the Wnt pathway as a possible mechanism of recurrence. To validate these findings and document whether Wnt inhibition may sensitize cells to chemotherapy, we analysed the expression of activated ß-catenin (and its downstream target BIRC5) using multiparameter phosphoflow cytometry and tested the efficacy of a recently developed Wnt inhibitor, iCRT14, in ALL cell lines and patient samples. We observed increased activation of ß-catenin at relapse in 6/10 patients. Furthermore, treatment of leukaemic cell lines with iCRT14 led to significant downregulation of Wnt target genes and combination with traditional chemotherapeutic drugs resulted in a synergistic decrease in viability as well as a significant increase in apoptotic cell death. Finally, pre-treatment of purified blasts from patients with relapsed leukaemia with the Wnt inhibitor followed by exposure to prednisolone, restored chemosensitivity in these cells. Our results demonstrate that overactivation of the Wnt pathway may contribute to chemoresistance in relapsed childhood ALL and that Wnt-inhibition may be a promising therapeutic approach.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Wnt Proteins/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Resistance, Neoplasm/genetics , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunophenotyping , Neoplasm Recurrence, Local , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects
14.
Proc Natl Acad Sci U S A ; 108(15): 5954-63, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21393571

ABSTRACT

Misregulated ß-catenin responsive transcription (CRT) has been implicated in the genesis of various malignancies, including colorectal carcinomas, and it is a key therapeutic target in combating various cancers. Despite significant effort, successful clinical implementation of CRT inhibitory therapeutics remains a challenging goal. This is, in part, because of the challenge of identifying inhibitory compounds that specifically modulate the nuclear transcriptional activity of ß-catenin while not affecting its cytoskeletal function in stabilizing adherens junctions at the cell membrane. Here, we report an RNAi-based modifier screening strategy for the identification of CRT inhibitors. Our data provide support for the specificity of these inhibitory compounds in antagonizing the transcriptional function of nuclear ß-catenin. We show that these inhibitors efficiently block Wnt/ß-catenin-induced target genes and phenotypes in various mammalian and cancer cell lines. Importantly, these Wnt inhibitors are specifically cytotoxic to human colon tumor biopsy cultures as well as colon cancer cell lines that exhibit deregulated Wnt signaling.


Subject(s)
Antineoplastic Agents/chemistry , Drug Discovery/methods , RNA Interference , Transcription, Genetic/drug effects , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster , Drug Screening Assays, Antitumor , Genes, Reporter , High-Throughput Screening Assays , Humans , Mice , Oxazoles/chemistry , Oxazoles/isolation & purification , Oxazoles/therapeutic use , Small Molecule Libraries , Wnt Proteins/genetics , Wnt1 Protein/antagonists & inhibitors , Wnt1 Protein/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics
15.
ACS Infect Dis ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38938101

ABSTRACT

A newly discovered E3 ubiquitin ligase, UBR7, plays a crucial role in histone H2BK120 monoubiquitination. Here, we report a novel function of UBR7 in promoting hepatitis B virus (HBV) pathogenesis, which further leads to HBV-induced hepatocellular carcinoma (HCC). Transcriptomics analysis from HCC patients revealed the deregulation of UBR7 in cancer. Remarkably, targeting UBR7, particularly its catalytic function, led to a significant decrease in viral copy numbers. We also identified the speckled family protein Sp110 as an important substrate of UBR7. Notably, Sp110 has been previously shown to be a resident of promyelocytic leukemia nuclear bodies (PML-NBs), where it remains SUMOylated, and during HBV infection, it undergoes deSUMOylation and exits the PML body. We observed that UBR7 ubiquitinates Sp110 at critical residues within its SAND domain. Sp110 ubiquitination downregulates genes in the type I interferon response pathway. Comparative analysis of RNA-Seq from the UBR7/Sp110 knockdown data set confirmed that the IFN-ß signaling pathway gets deregulated in HCC cells in the presence of HBV. Single-cell RNA-Seq analysis of patient samples further confirmed the inverse correlation between the expression of Sp110/UBR7 and the inflammation score. Notably, silencing of UBR7 induces IRF7 phosphorylation, thereby augmenting interferon (IFN)-ß and the downstream interferon-stimulated genes (ISGs). Further, wild-type but not the ubiquitination-defective mutant of Sp110 could be recruited to the type I interferon response pathway genes. Our study establishes a new function of UBR7 in non-histone protein ubiquitination, promoting viral persistence, and has important implications for the development of therapeutic strategies targeting HBV-induced HCC.

16.
Bioeng Transl Med ; 9(2): e10628, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38435825

ABSTRACT

We present a novel framework combining single-cell phenotypic data with single-cell transcriptomic analysis to identify factors underpinning heterogeneity in antitumor immune response. We developed a pairwise, tumor-immune discretized interaction assay between natural killer (NK-92MI) cells and patient-derived head and neck squamous cell carcinoma (HNSCC) cell lines on a microfluidic cell-trapping platform. Furthermore we generated a deep-learning computer vision algorithm that is capable of automating the acquisition and analysis of a large, live-cell imaging data set (>1 million) of paired tumor-immune interactions spanning a time course of 24 h across multiple HNSCC lines (n = 10). Finally, we combined the response data measured by Kaplan-Meier survival analysis against NK-mediated killing with downstream single-cell transcriptomic analysis to interrogate molecular signatures associated with NK-effector response. As proof-of-concept for the proposed framework, we efficiently identified MHC class I-driven cytotoxic resistance as a key mechanism for immune evasion in nonresponders, while enhanced expression of cell adhesion molecules was found to be correlated with sensitivity against NK-mediated cytotoxicity. We conclude that this integrated, data-driven phenotypic approach holds tremendous promise in advancing the rapid identification of new mechanisms and therapeutic targets related to immune evasion and response.

17.
Cell Rep ; 43(7): 114400, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38935501

ABSTRACT

ADAR1-mediated RNA editing establishes immune tolerance to endogenous double-stranded RNA (dsRNA) by preventing its sensing, primarily by MDA5. Although deleting Ifih1 (encoding MDA5) rescues embryonic lethality in ADAR1-deficient mice, they still experience early postnatal death, and removing other MDA5 signaling proteins does not yield the same rescue. Here, we show that ablation of MDA5 in a liver-specific Adar knockout (KO) murine model fails to rescue hepatic abnormalities caused by ADAR1 loss. Ifih1;Adar double KO (dKO) hepatocytes accumulate endogenous dsRNAs, leading to aberrant transition to a highly inflammatory state and recruitment of macrophages into dKO livers. Mechanistically, progranulin (PGRN) appears to mediate ADAR1 deficiency-induced liver pathology, promoting interferon signaling and attracting epidermal growth factor receptor (EGFR)+ macrophages into dKO liver, exacerbating hepatic inflammation. Notably, the PGRN-EGFR crosstalk communication and consequent immune responses are significantly repressed in ADAR1high tumors, revealing that pre-neoplastic or neoplastic cells can exploit ADAR1-dependent immune tolerance to facilitate immune evasion.


Subject(s)
Adenosine Deaminase , ErbB Receptors , Hepatocytes , Interferon-Induced Helicase, IFIH1 , Liver , Macrophages , Mice, Knockout , Progranulins , Animals , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , ErbB Receptors/metabolism , Macrophages/metabolism , Macrophages/immunology , Progranulins/metabolism , Progranulins/genetics , Liver/metabolism , Liver/immunology , Liver/pathology , Hepatocytes/metabolism , Mice , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Signal Transduction , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice, Inbred C57BL , RNA, Double-Stranded/metabolism , RNA Editing
18.
Metabolism ; 152: 155744, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38029839

ABSTRACT

BACKGROUND: High body mass index (BMI) is a major risk factor for cancer development, but its impact on the global burden of cancer remains unclear. METHODS: We estimated global and regional temporal trends in the burden of cancer attributable to high BMI, and the contributions of various cancer types using the framework of the Global Burden of Disease Study. RESULTS: From 2010 to 2019, there was a 35 % increase in deaths and a 34 % increase in disability-adjusted life-years from cancers attributable to high BMI. The age-standardized death rates for cancer attributable to high BMI increased over the study period (annual percentage change [APC] +0.48 %, 95 % CI 0.22 to 0.74 %). The greatest number of deaths from cancer attributable to high BMI occurred in Europe, but the fastest-growing age-standardized death rates and disability-adjusted life-years occurred in Southeast Asia. Liver cancer was the fastest-growing cause of cancer mortality (APC: 1.37 %, 95 % CI 1.25 to 1.49 %) attributable to high BMI. CONCLUSION: The global burden of cancer-related deaths attributable to high BMI has increased substantially from 2010 to 2019. The greatest increase in age-standardized death rates occurred in Southeast Asia, and liver cancer is the fastest-growing cause of cancer mortality attributable to high BMI. Urgent and sustained measures are required at a global and regional level to reverse these trends and slow the growing burden of cancer attributed to high BMI.


Subject(s)
Liver Neoplasms , Humans , Body Mass Index , Quality-Adjusted Life Years , Risk Factors , Europe/epidemiology
19.
Nat Cancer ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844817

ABSTRACT

Many individuals with cancer are resistant to immunotherapies. Here, we identify the gene encoding the pyrimidine salvage pathway enzyme cytidine deaminase (CDA) among the top upregulated metabolic genes in several immunotherapy-resistant tumors. We show that CDA in cancer cells contributes to the uridine diphosphate (UDP) pool. Extracellular UDP hijacks immunosuppressive tumor-associated macrophages (TAMs) through its receptor P2Y6. Pharmacologic or genetic inhibition of CDA in cancer cells (or P2Y6 in TAMs) disrupts TAM-mediated immunosuppression, promoting cytotoxic T cell entry and susceptibility to anti-programmed cell death protein 1 (anti-PD-1) treatment in resistant pancreatic ductal adenocarcinoma (PDAC) and melanoma models. Conversely, CDA overexpression in CDA-depleted PDACs or anti-PD-1-responsive colorectal tumors or systemic UDP administration (re)establishes resistance. In individuals with PDAC, high CDA levels in cancer cells correlate with increased TAMs, lower cytotoxic T cells and possibly anti-PD-1 resistance. In a pan-cancer single-cell atlas, CDAhigh cancer cells match with T cell cytotoxicity dysfunction and P2RY6high TAMs. Overall, we suggest CDA and P2Y6 as potential targets for cancer immunotherapy.

20.
Cancer Discov ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38885349

ABSTRACT

Over-consumption of iron-rich red meat and hereditary or genetic iron overload are associated with increased risk of colorectal carcinogenesis, yet the mechanistic basis of how metal-mediated signaling leads to oncogenesis remains enigmatic. Using fresh colorectal cancer (CRC) samples we identify Pirin, an iron sensor, that overcomes a rate-limiting step in oncogenesis, by re-activating the dormant human-reverse-transcriptase (hTERT) subunit of telomerase holoenzyme in an iron-(Fe3+)-dependent-manner and thereby drives CRCs. Chemical genetic screens combined with isothermal-dose response fingerprinting and mass-spectrometry identified a small molecule SP2509, that specifically inhibits Pirin-mediated hTERT reactivation in CRCs by competing with iron-(Fe3+) binding. Our findings, first to document how metal ions reactivate telomerase, provide a molecular mechanism for the well-known association between red meat, and increased incidence of CRCs. Small molecules like SP2509 represent a novel modality to target telomerase that acts as driver of 90% human cancers and is yet to be targeted in clinic.

SELECTION OF CITATIONS
SEARCH DETAIL