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










Publication year range
1.
Lancet Haematol ; 11(2): e136-e146, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38190832

ABSTRACT

BACKGROUND: Diffuse large B-cell lymphoma comprises nearly 30% of non-Hodgkin lymphoma cases and patients with relapsed or refractory diffuse large B-cell lymphoma who are ineligible for stem-cell transplantation have few treatment options and poor prognoses. We aimed to determine whether the novel combination of polatuzumab vedotin in combination with rituximab and lenalidomide (Pola+R+Len) would provide a tolerable treatment option with enhanced antitumour response in patients with relapsed or refractory diffuse large B-cell lymphoma. METHODS: This completed phase 1b/2, open-label, multicentre, single-arm study (GO29834) evaluated the safety and efficacy of Pola+R+Len in patients with relapsed or refractory diffuse large B-cell lymphoma at 19 sites in three countries (USA, Spain, and UK). Patients (≥18 years old) were eligible for inclusion if they had histologically documented CD20-positive relapsed or refractory diffuse large B-cell lymphoma and Eastern Cooperative Oncology Group performance status of 2 or lower, had received at least one previous line of chemoimmunotherapy, including an anti-CD20 agent, and were ineligible for stem-cell transplantation. The dose-escalation phase (1b) used escalating doses of lenalidomide to find the recommended phase 2 dose. Patients received six 28-day cycles of induction treatment with intravenous rituximab 375 mg/m2 and intravenous polatuzumab vedotin 1·8 mg/kg (all cohorts) plus oral lenalidomide at the following doses: 10 mg (cohort A); 15 mg (cohort B); and 20 mg (cohort C). Rituximab and polatuzumab vedotin were administered on day 1 and lenalidomide on days 1-21 of each 28-day cycle. During the dose-expansion phase (2), patients received six 28-day cycles of Pola+R+Len at the recommended phase 2 dose established during dose escalation. In both phases, patients with a complete response or partial response at the end of induction were eligible for post-induction therapy with rituximab 375 mg/m2 on day 1 and lenalidomide 10 mg/day on days 1-21 of each 28-day cycle for a maximum of 6 cycles. The primary safety objective of the dose-escalation phase was identification of the maximum tolerated dose through incidence of dose-limiting toxic effects. The primary efficacy outcome of the dose-expansion phase was Independent Review Committee-assessed complete response rate at end of induction, based on PET-CT. Analyses were conducted in the safety population, which included all patients who received at least one dose of any study drug, and the efficacy population, which included all patients who received at least one dose of any study drug at the recommended phase 2 dose. This study is registered with ClinicalTrials.gov, number NCT02600897. FINDINGS: Between July 11, 2017 and Feb 3, 2020, 57 patients were enrolled (median age 71 years [IQR 60-75]; 38 [67%] were male and 19 (33%) were female; 47 [82%] were not Hispanic or Latino; and the median previous lines of therapy was 2 [IQR 1-3]). 18 participants were included in phase 1b and 39 were included in phase 2. Phase 1b confirmed a 20 mg recommended phase 2 dose for lenalidomide. After a median follow-up of 11·8 months (IQR 4·7-25·8), the complete response rate, as assessed by the Independent Review Committee, was 31% (90% CI 20-43). The most common grade 3-4 adverse events were neutropenia (35 [61%] of 57) and thrombocytopenia (eight [14%] of 57). Serious adverse events were reported in 23 (40%) of 57 patients and one patient died due to a treatment-related adverse event (neutropenic sepsis). INTERPRETATION: Although the combination of Pola+R+Len did not meet the prespecified activity threshold, some patients derived clinical benefit and the regimen had a tolerable safety profile in patients with relapsed or refractory diffuse large B-cell lymphoma. FUNDING: Genentech/F Hoffmann-La Roche.


Subject(s)
Antibodies, Monoclonal , Immunoconjugates , Lymphoma, Large B-Cell, Diffuse , Lymphoma, Non-Hodgkin , Neutropenia , Humans , Male , Female , Aged , Adolescent , Rituximab/adverse effects , Lenalidomide/therapeutic use , Positron Emission Tomography Computed Tomography , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Neutropenia/etiology
3.
Nat Commun ; 14(1): 2147, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37072421

ABSTRACT

Data on long-term outcomes and biological drivers associated with depth of remission after BCL2 inhibition by venetoclax in the treatment of chronic lymphocytic leukemia (CLL) are limited. In this open-label parallel-group phase-3 study, 432 patients with previously untreated CLL were randomized (1:1) to receive either 1-year venetoclax-obinutuzumab (Ven-Obi, 216 patients) or chlorambucil-Obi (Clb-Obi, 216 patients) therapy (NCT02242942). The primary endpoint was investigator-assessed progression-free survival (PFS); secondary endpoints included minimal residual disease (MRD) and overall survival. RNA sequencing of CD19-enriched blood was conducted for exploratory post-hoc analyses. After a median follow-up of 65.4 months, PFS is significantly superior for Ven-Obi compared to Clb-Obi (Hazard ratio [HR] 0.35 [95% CI 0.26-0.46], p < 0.0001). At 5 years after randomization, the estimated PFS rate is 62.6% after Ven-Obi and 27.0% after Clb-Obi. In both arms, MRD status at the end of therapy is associated with longer PFS. MRD + ( ≥ 10-4) status is associated with increased expression of multi-drug resistance gene ABCB1 (MDR1), whereas MRD6 (< 10-6) is associated with BCL2L11 (BIM) expression. Inflammatory response pathways are enriched in MRD+ patient solely in the Ven-Obi arm. These data indicate sustained long-term efficacy of fixed-duration Ven-Obi in patients with previously untreated CLL. The distinct transcriptomic profile of MRD+ status suggests possible biological vulnerabilities.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Transcriptome , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chlorambucil/therapeutic use , Chlorambucil/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
4.
Cell Mol Immunol ; 20(3): 277-291, 2023 03.
Article in English | MEDLINE | ID: mdl-36631557

ABSTRACT

Upon recognition of foreign antigens, naïve B cells undergo rapid activation, growth, and proliferation. How B-cell growth and proliferation are coupled with activation remains poorly understood. Combining CRISPR/Cas9-mediated functional analysis and mouse genetics approaches, we found that Dhx33, an activation-induced RNA helicase, plays a critical role in coupling B-cell activation with growth and proliferation. Mutant mice with B-cell-specific deletion of Dhx33 exhibited impaired B-cell development, germinal center reactions, plasma cell differentiation, and antibody production. Dhx33-deficient B cells appeared normal in the steady state and early stage of activation but were retarded in growth and proliferation. Mechanistically, Dhx33 played an indispensable role in activation-induced upregulation of ribosomal DNA (rDNA) transcription. In the absence of Dhx33, activated B cells were compromised in their ability to ramp up 47S ribosomal RNA (rRNA) production and ribosome biogenesis, resulting in nucleolar stress, p53 accumulation, and cellular death. Our findings demonstrate an essential role for Dhx33 in coupling B-cell activation with growth and proliferation and suggest that Dhx33 inhibition is a potential therapy for lymphoma and antibody-mediated autoimmune diseases.


Subject(s)
RNA, Ribosomal , Animals , Mice , Cell Cycle , Cell Proliferation , RNA, Ribosomal/genetics , Up-Regulation
5.
Cells ; 9(1)2020 01 20.
Article in English | MEDLINE | ID: mdl-31968623

ABSTRACT

We present data showing that Iodotyrosine Deiodinase (IYD) is a dual-function enzyme acting as a catalyst in metabolism and a receptor for cooperative stem cell differentiation. IYD is present both in thyroid cells where it is critical for scavenging iodine from halogenated by-products of thyroid hormone production and on hematopoietic stem cells. To close the cooperative loop, the mono- and di-Iodotyrosine (MIT and DIT) substrates of IYD in the thyroid are also agonists for IYD now acting as a receptor on bone marrow stem cells. While studying intracellular combinatorial antibody libraries, we discovered an agonist antibody, H3 Ab, of which the target is the enzyme IYD. When agonized by H3 Ab, IYD expressed on stem cells induces differentiation of the cells into brown adipocyte-like cells, which selectively migrate to mouse heart tissue. H3 Ab also binds to IYD expressed on human myocardium. Thus, one has a single enzyme acting in different ways on different cells for the cooperative purpose of enhancing thermogenesis or of regenerating damaged heart tissue.


Subject(s)
Adipocytes, Brown/cytology , Antibodies/pharmacology , Cell Movement , Myocardium/cytology , Stem Cells/cytology , Adipocytes, Brown/drug effects , Adipocytes, Brown/ultrastructure , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Myocardium/ultrastructure , Stem Cells/drug effects
6.
SLAS Discov ; 25(2): 215-222, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31849250

ABSTRACT

The real-time quantification of target engagement (TE) by small-molecule ligands in living cells remains technically challenging. Systematic quantification of such interactions in a high-throughput setting holds promise for identification of target-specific, potent small molecules within a pathophysiological and biologically relevant cellular context. The salt-inducible kinases (SIKs) belong to a subfamily of the AMP-activated protein kinase (AMPK) family and are composed of three isoforms in humans (SIK1, SIK2, and SIK3). They modulate the production of pro- and anti-inflammatory cytokines in immune cells. Although pan-SIK inhibitors are sufficient to reverse SIK-dependent inflammatory responses, the apparent toxicity associated with SIK3 inhibition suggests that isoform-specific inhibition is required to realize therapeutic benefit with acceptable safety margins. Here, we used the NanoBRET TE intracellular kinase assay, a sensitive energy transfer technique, to directly measure molecular proximity and quantify TE in HEK293T cells overexpressing SIK2 or SIK3. Our 384-well high-throughput screening of 530 compounds demonstrates that the NanoBRET TE intracellular kinase assay was sensitive and robust enough to reveal differential engagement of candidate compounds with the two SIK isoforms and further highlights the feasibility of high-throughput implementation of NanoBRET TE intracellular kinase assays for target-driven small-molecule screening.


Subject(s)
Phosphotransferases/isolation & purification , Protein Isoforms/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fluorescence Resonance Energy Transfer , Gene Expression Regulation, Enzymologic/drug effects , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Phosphotransferases/genetics , Protein Isoforms/antagonists & inhibitors , Protein Kinases/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors
7.
Nat Commun ; 10(1): 3496, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31375680

ABSTRACT

The timely mobilization of hematopoietic stem and progenitor cells (HSPCs) is essential for maintaining hematopoietic and tissue leukocyte homeostasis. Understanding how HSPCs migrate between bone marrow (BM) and peripheral tissues is of great significance in the clinical setting, where therapeutic strategies for modulating their migration capacity determine the clinical outcome. Here, we identify an epigenetic regulator, Phc2, as a critical modulator of HSPC trafficking. The genetic ablation of Phc2 in mice causes a severe defect in HSPC mobilization through the derepression of Vcam1 in bone marrow stromal cells (BMSCs), ultimately leading to a systemic immunodeficiency. Moreover, the pharmacological inhibition of VCAM-1 in Phc2-deficient mice reverses the symptoms. We further determine that Phc2-dependent Vcam1 repression in BMSCs is mediated by the epigenetic regulation of H3K27me3 and H2AK119ub. Together, our data demonstrate a cell-extrinsic role for Phc2 in controlling the mobilization of HSPCs by finely tuning their bone marrow niche.


Subject(s)
Cell Movement/genetics , Epigenetic Repression , Hematopoietic Stem Cells/immunology , Polycomb Repressive Complex 2/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Animals , Bone Marrow Transplantation/adverse effects , Cell Movement/immunology , Cells, Cultured , DNA Methylation/immunology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/adverse effects , Histones/genetics , Histones/metabolism , Mice , Mice, Knockout , Models, Animal , Polycomb Repressive Complex 2/genetics , Primary Cell Culture , Vascular Cell Adhesion Molecule-1/antagonists & inhibitors
8.
Nat Med ; 25(2): 301-311, 2019 02.
Article in English | MEDLINE | ID: mdl-30643286

ABSTRACT

Cancer cells develop mechanisms to escape immunosurveillance, among which modulating the expression of immune suppressive messenger RNAs is most well-documented. However, how this is molecularly achieved remains largely unresolved. Here, we develop an in vivo mouse model of liver cancer to study oncogene cooperation in immunosurveillance. We show that MYC overexpression (MYCTg) synergizes with KRASG12D to induce an aggressive liver tumor leading to metastasis formation and reduced mouse survival compared with KRASG12D alone. Genome-wide ribosomal footprinting of MYCTg;KRASG12 tumors compared with KRASG12D revealed potential alterations in translation of mRNAs, including programmed-death-ligand 1 (PD-L1). Further analysis revealed that PD-L1 translation is repressed in KRASG12D tumors by functional, non-canonical upstream open reading frames in its 5' untranslated region, which is bypassed in MYCTg;KRASG12D tumors to evade immune attack. We show that this mechanism of PD-L1 translational upregulation was effectively targeted by a potent, clinical compound that inhibits eIF4E phosphorylation, eFT508, which reverses the aggressive and metastatic characteristics of MYCTg;KRASG12D tumors. Together, these studies reveal how immune-checkpoint proteins are manipulated by distinct oncogenes at the level of mRNA translation, which can be exploited for new immunotherapies.


Subject(s)
Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Protein Biosynthesis , 5' Untranslated Regions/genetics , Animals , B7-H1 Antigen/metabolism , Base Sequence , Disease Progression , Down-Regulation , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic , Immune Evasion , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice, Inbred C57BL , Neoplasm Metastasis , Open Reading Frames/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Transcription, Genetic , Tumor Microenvironment , Up-Regulation/genetics
9.
Methods Mol Biol ; 1712: 1-18, 2018.
Article in English | MEDLINE | ID: mdl-29224064

ABSTRACT

Recent advances in global translatome analysis technologies enable us to understand how translational regulation of gene expression modulates cellular functions. In this chapter, we present an integrated method to measure various aspects of translatome by polysome profiling and ribosome profiling using purified B cells. We standardized our protocols to directly compare the results from these two approaches. Parallel assessment of translatome with these two approaches can generate a comprehensive picture on how translational regulation determines protein output.


Subject(s)
Gene Expression Profiling/methods , Polyribosomes/genetics , Protein Biosynthesis/genetics , Ribosomes/genetics , Animals , B-Lymphocytes/metabolism , Models, Animal , Proteins/genetics , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Ribosomes/chemistry , Sequence Analysis, RNA
10.
PLoS Genet ; 13(2): e1006623, 2017 02.
Article in English | MEDLINE | ID: mdl-28241004

ABSTRACT

MicroRNAs (miRNAs) are thought to exert their functions by modulating the expression of hundreds of target genes and each to a small degree, but it remains unclear how small changes in hundreds of target genes are translated into the specific function of a miRNA. Here, we conducted an integrated analysis of transcriptome and translatome of primary B cells from mutant mice expressing miR-17~92 at three different levels to address this issue. We found that target genes exhibit differential sensitivity to miRNA suppression and that only a small fraction of target genes are actually suppressed by a given concentration of miRNA under physiological conditions. Transgenic expression and deletion of the same miRNA gene regulate largely distinct sets of target genes. miR-17~92 controls target gene expression mainly through translational repression and 5'UTR plays an important role in regulating target gene sensitivity to miRNA suppression. These findings provide molecular insights into a model in which miRNAs exert their specific functions through a small number of key target genes.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Protein Biosynthesis/genetics , Transcriptome/genetics , 5' Untranslated Regions/genetics , Animals , B-Lymphocytes/cytology , Base Sequence , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cells, Cultured , Flow Cytometry , Gene Expression Profiling/methods , Immunoblotting , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomes/genetics , Ribosomes/metabolism
11.
Nat Commun ; 7: 12207, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27481093

ABSTRACT

The molecular mechanisms that regulate B-cell development and tolerance remain incompletely understood. In this study, we identify a critical role for the miR-17∼92 microRNA cluster in regulating B-cell central tolerance and demonstrate that these miRNAs control early B-cell development in a cell-intrinsic manner. While the cluster member miR-19 suppresses the expression of Pten and plays a key role in regulating B-cell tolerance, miR-17 controls early B-cell development through other molecular pathways. These findings demonstrate differential control of two closely linked B-cell developmental stages by different members of a single microRNA cluster through distinct molecular pathways.


Subject(s)
B-Lymphocytes/physiology , Immune Tolerance/genetics , Lymphocyte Activation/genetics , MicroRNAs/physiology , PTEN Phosphohydrolase/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal
12.
J Exp Med ; 213(9): 1901-19, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27481129

ABSTRACT

MicroRNA (miRNA) deficiency impairs the generation of T follicular helper (Tfh) cells, but the contribution of individual miRNAs to this phenotype remains poorly understood. In this study, we performed deep sequencing analysis of miRNAs expressed in Tfh cells and identified a five-miRNA signature. Analyses of mutant mice deficient of these miRNAs revealed that miR-22 and miR-183/96/182 are dispensable, but miR-155 is essential for the generation and function of Tfh cells. miR-155 deficiency led to decreased proliferation specifically at the late stage of Tfh cell differentiation and reduced CD40 ligand (CD40L) expression on antigen-specific CD4(+) T cells. Mechanistically, miR-155 repressed the expression of Peli1, a ubiquitin ligase that promotes the degradation of the NF-κB family transcription factor c-Rel, which controls cellular proliferation and CD40L expression. Therefore, our study identifies a novel miR-155-Peli1-c-Rel pathway that specifically regulates Tfh cell generation and function.


Subject(s)
MicroRNAs/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-rel/physiology , Signal Transduction/physiology , T-Lymphocytes, Helper-Inducer/immunology , Ubiquitin-Protein Ligases/physiology , Animals , CD40 Ligand/analysis , Cell Differentiation , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , T-Lymphocytes, Cytotoxic/immunology
13.
Immunity ; 44(6): 1284-98, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332731

ABSTRACT

T helper 17 (Th17) cells are key players in autoimmune diseases. However, the roles of non-coding RNAs in Th17 cell development and function are largely unknown. We found that deletion of the endoribonuclease-encoding Dicer1 specifically in Th17 cells protected mice from experimental autoimmune encephalomyelitis. We found that the Dicer1-regulated microRNA (miR)-183-96-182 cluster (miR-183C) was highly expressed in Th17 cells and was induced by cytokine IL-6-STAT3 signaling. miR-183C expression enhanced pathogenic cytokine production from Th17 cells during their development and promoted autoimmunity. Mechanistically, miR-183C in Th17 cells directly repressed expression of the transcription factor Foxo1. Foxo1 negatively regulated the pathogenicity of Th17 cells in part by inhibiting expression of cytokine receptor IL-1R1. These findings indicate that the miR-183C drives Th17 pathogenicity in autoimmune diseases via inhibition of Foxo1 and present promising therapeutic targets.


Subject(s)
DEAD-box RNA Helicases/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Forkhead Box Protein O1/metabolism , MicroRNAs/genetics , Multiple Sclerosis/immunology , Ribonuclease III/metabolism , Th17 Cells/physiology , Animals , Cells, Cultured , DEAD-box RNA Helicases/genetics , Forkhead Box Protein O1/genetics , Humans , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1 Type I/metabolism , Ribonuclease III/genetics , STAT3 Transcription Factor/metabolism
14.
15.
Front Genet ; 6: 340, 2015.
Article in English | MEDLINE | ID: mdl-26697058

ABSTRACT

Transient transfection of chemically synthesized microRNA (miRNA) mimics is being used extensively to study the functions and mechanisms of endogenous miRNAs. However, it remains unclear whether transfected miRNAs behave similarly to endogenous miRNAs. Here we show that transient transfection of miRNA mimics into HeLa cells by a commonly used method led to the accumulation of high molecular weight RNA species and a few hundred fold increase in mature miRNA levels. In contrast, expression of the same miRNAs through lentiviral infection or plasmid transfection of HeLa cells, transgenic expression in primary lymphocytes, and endogenous overexpression in lymphoma and leukemia cell lines did not lead to the appearance of high molecular weight RNA species. The increase of mature miRNA levels in these cells was below 10-fold, which was sufficient to suppress target gene expression and to drive lymphoma development in mice. Moreover, transient transfection of miRNA mimics at high concentrations caused non-specific alterations in gene expression, while at low concentrations achieved expression levels comparable to other methods but failed to efficiently suppress target gene expression. Small RNA deep sequencing analysis revealed that the guide strands of miRNA mimics were frequently mutated, while unnatural passenger strands of some miRNA mimics accumulated to high levels. The high molecular weight RNA species were a heterogeneous mixture of several classes of RNA species generated by concatemerization, 5'- and 3'-end tailing of miRNA mimics. We speculate that the supraphysiological levels of mature miRNAs and these artifactual RNA species led to non-specific changes in gene expression. Our results have important implications for the design and interpretation of experiments primarily employing transient transfection of miRNA mimics.

16.
Elife ; 4: e07072, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26111384

ABSTRACT

The Reproducibility Project: Cancer Biology seeks to address growing concerns about reproducibility in scientific research by replicating selected results from a substantial number of high-profile papers in the field of cancer biology published between 2010 and 2012. This Registered report describes the proposed replication plan of key experiments from 'BET bromodomain inhibition as a therapeutic strategy to target c-Myc' by Delmore and colleagues, published in Cell in 2011 (Delmore et al., 2011). The key experiments that will be replicated are those reported in Figures 3B and 7C-E. Delmore and colleagues demonstrated that treatment with JQ1, a small molecular inhibitor targeting BET bromodomains, resulted in the transcriptional down-regulation of the c-Myc oncogene in vitro (Figure 3B; Delmore et al., 2011). To assess the therapeutic efficacy of JQ1 in vivo, mice bearing multiple myeloma (MM) lesions were treated with JQ1 before evaluation for tumor burden and overall survival. JQ1 treatment significantly reduced disease burden and increased survival time (Figure 7C-E; Delmore et al., 2011). The Reproducibility Project: Cancer Biology is a collaboration between the Center for Open Science and Science Exchange and the results of the replications will be published in eLife.


Subject(s)
Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Azepines/metabolism , Azepines/therapeutic use , Disease Models, Animal , Down-Regulation , Gene Expression Regulation/drug effects , Mice , Multiple Myeloma/pathology , Survival Analysis , Treatment Outcome , Triazoles/metabolism , Triazoles/therapeutic use
18.
EMBO J ; 32(17): 2377-91, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23921550

ABSTRACT

MicroRNAs (miRNAs) have been broadly implicated in cancer, but their exact function and mechanism in carcinogenesis remain poorly understood. Elevated miR-17~92 expression is frequently found in human cancers, mainly due to gene amplification and Myc-mediated transcriptional upregulation. Here we show that B cell-specific miR-17~92 transgenic mice developed lymphomas with high penetrance and that, conversely, Myc-driven lymphomagenesis stringently requires two intact alleles of miR-17~92. We experimentally identified miR-17~92 target genes by PAR-CLIP and validated select target genes in miR-17~92 transgenic mice. These analyses demonstrate that miR-17~92 drives lymphomagenesis by suppressing the expression of multiple negative regulators of the PI3K and NFκB pathways and by inhibiting the mitochondrial apoptosis pathway. Accordingly, miR-17~92-driven lymphoma cells exhibited constitutive activation of the PI3K and NFκB pathways and chemical inhibition of either pathway reduced tumour size and prolonged the survival of lymphoma-bearing mice. These findings establish miR-17~92 as a powerful cancer driver that coordinates the activation of multiple oncogenic pathways, and demonstrate for the first time that chemical inhibition of miRNA downstream pathways has therapeutic value in treating cancers caused by miRNA dysregulation.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma/genetics , MicroRNAs/physiology , Animals , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Cell Proliferation , Cell Survival/genetics , Homeodomain Proteins/genetics , Humans , Imidazoles/pharmacology , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morpholines/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/administration & dosage , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Quinoxalines/pharmacology , RNA, Long Noncoding , Reproducibility of Results
19.
Nat Immunol ; 14(8): 849-57, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23812097

ABSTRACT

Follicular helper T cells (T(FH) cells) provide critical help to B cells during humoral immune responses. Here we report that mice with T cell-specific deletion of the miR-17∼92 family of microRNAs (miRNAs) had substantially compromised T(FH) differentiation, germinal-center formation and antibody responses and failed to control chronic viral infection. Conversely, mice with T cell-specific expression of a transgene encoding miR-17∼92 spontaneously accumulated T(FH) cells and developed a fatal immunopathology. Mechanistically, the miR-17∼92 family controlled the migration of CD4(+) T cells into B cell follicles by regulating signaling intensity from the inducible costimulator ICOS and kinase PI(3)K by suppressing expression of the phosphatase PHLPP2. Our findings demonstrate an essential role for the miR-17∼92 family in T(FH) differentiation and establish PHLPP2 as an important mediator of their function in this process.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Germinal Center/immunology , MicroRNAs/immunology , Nuclear Proteins/immunology , Phosphoprotein Phosphatases/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Flow Cytometry , Germinal Center/cytology , Immunity, Humoral/immunology , Immunohistochemistry , Inducible T-Cell Co-Stimulator Protein/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol 3-Kinases/immunology , Signal Transduction/immunology , Specific Pathogen-Free Organisms , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/enzymology
20.
J Virol ; 86(2): 1109-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22090108

ABSTRACT

Replication and transcription activator (RTA), an immediate-early gene, is a key molecular switch to evoke lytic replication of gammaherpesviruses. Open reading frame 49 (ORF49) is conserved among gammaherpesviruses and shown to cooperate with RTA in regulating virus lytic replication. Here we show a molecular mechanism and in vivo functions of murine gammaherpesvirus 68 (MHV-68 or γHV-68) ORF49. MHV-68 ORF49 was transcribed and translated as a late gene. The ORF49 protein was associated with a virion, interacting with the ORF64 large tegument protein and the ORF25 capsid protein. Moreover, ORF49 directly bound to RTA and its negative cellular regulator, poly(ADP-ribose) polymerase-1 (PARP-1), and disrupted the interactions of RTA and PARP-1. Productive replication of an ORF49-deficient mutant virus (49S) was attenuated in vivo as well as in vitro. Likewise, latent infection was also impaired in the spleen of 49S-infected mice. Taken together, our results suggest that the virion-associated ORF49 protein may promote virus replication both in vitro and in vivo by providing an optimal environment in the early phase of virus infection as a derepressor of RTA.


Subject(s)
Gammaherpesvirinae/physiology , Gene Expression Regulation, Viral , Herpesviridae Infections/veterinary , Immediate-Early Proteins/genetics , Trans-Activators/metabolism , Viral Proteins/metabolism , Virion/physiology , Virus Replication , Animals , Cell Line , Cricetinae , Gammaherpesvirinae/genetics , Herpesviridae Infections/virology , Humans , Immediate-Early Proteins/metabolism , Mice , Open Reading Frames , Rodent Diseases/virology , Trans-Activators/genetics , Transcription, Genetic , Viral Proteins/genetics , Virion/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...