Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 75
1.
Cancer Discov ; 13(12): 2505-2506, 2023 12 12.
Article En | MEDLINE | ID: mdl-38084094

SUMMARY: The study by Bercier and colleagues investigates the mechanisms of action of arsenic trioxide (ATO). The authors find that ATO promotes transition of PML nuclear bodies to a gel-like state via the PML trimerization domain and a critical cysteine residue. Overall, this work sheds new light onto how PML-RARα, the oncogene of APL, is targeted by ATO for disease eradication. See related article by Bercier et al., p. 2548 (6).


Antineoplastic Agents , Arsenicals , Leukemia, Promyelocytic, Acute , Humans , Arsenic Trioxide/pharmacology , Arsenic Trioxide/therapeutic use , Arsenicals/pharmacology , Arsenicals/therapeutic use , Oxides/pharmacology , Oxides/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Oncogene Proteins, Fusion/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Cell Death Differ ; 2023 Oct 12.
Article En | MEDLINE | ID: mdl-37828086

Identification of mutations in histones in a number of human neoplasms and developmental syndromes represents the most compelling evidence to date for a causal role of epigenetic perturbations in human disease. In most cases, these mutations have gain of function properties that cause deviation from normal developmental processes leading to embryo defects and/or neoplastic transformation. These exciting discoveries represent a step-change in our understanding of the role of chromatin (dys)regulation in development and disease. However, the mechanisms of action of oncogenic histone mutations (oncohistones) remain only partially understood. Here, we critically assess existing literature on oncohistones focussing mainly on bone neoplasms. We show how it is possible to draw parallels with some of the cell-autonomous mechanisms of action described in paediatric brain cancer, although the functions of oncohistones in bone tumours remain under-investigated. In this respect, it is becoming clear that histone mutations targeting the same residues display, at least in part, tissue-specific oncogenic mechanisms. Furthermore, it is emerging that cancer cells carrying oncohistones can modify the surrounding microenvironment to support growth and/or alter differentiation trajectories. A better understanding of oncohistone function in different neoplasms provide potential for identification of signalling that could be targeted therapeutically. Finally, we discuss some of the main concepts and future directions in this research area, while also drawing possible connections and parallels with other cancer epigenetic mechanisms.

3.
Nat Commun ; 13(1): 6830, 2022 11 11.
Article En | MEDLINE | ID: mdl-36369285

Current concepts regarding the biology of aging are primarily based on studies aimed at identifying factors regulating lifespan. However, lifespan as a sole proxy measure for aging can be of limited value because it may be restricted by specific pathologies. Here, we employ large-scale phenotyping to analyze hundreds of markers in aging male C57BL/6J mice. For each phenotype, we establish lifetime profiles to determine when age-dependent change is first detectable relative to the young adult baseline. We examine key lifespan regulators (putative anti-aging interventions; PAAIs) for a possible countering of aging. Importantly, unlike most previous studies, we include in our study design young treated groups of animals, subjected to PAAIs prior to the onset of detectable age-dependent phenotypic change. Many PAAI effects influence phenotypes long before the onset of detectable age-dependent change, but, importantly, do not alter the rate of phenotypic change. Hence, these PAAIs have limited effects on aging.


Aging , Longevity , Mice , Animals , Male , Longevity/genetics , Mice, Inbred C57BL , Aging/physiology , Phenotype
4.
EMBO J ; 41(23): e110595, 2022 12 01.
Article En | MEDLINE | ID: mdl-36305367

Mammalian SWI/SNF/BAF chromatin remodeling complexes influence cell lineage determination. While the contribution of these complexes to neural progenitor cell (NPC) proliferation and differentiation has been reported, little is known about the transcriptional profiles that determine neurogenesis or gliogenesis. Here, we report that BCL7A is a modulator of the SWI/SNF/BAF complex that stimulates the genome-wide occupancy of the ATPase subunit BRG1. We demonstrate that BCL7A is dispensable for SWI/SNF/BAF complex integrity, whereas it is essential to regulate Notch/Wnt pathway signaling and mitochondrial bioenergetics in differentiating NPCs. Pharmacological stimulation of Wnt signaling restores mitochondrial respiration and attenuates the defective neurogenic patterns observed in NPCs lacking BCL7A. Consistently, treatment with an enhancer of mitochondrial biogenesis, pioglitazone, partially restores mitochondrial respiration and stimulates neuronal differentiation of BCL7A-deficient NPCs. Using conditional BCL7A knockout mice, we reveal that BCL7A expression in NPCs and postmitotic neurons is required for neuronal plasticity and supports behavioral and cognitive performance. Together, our findings define the specific contribution of BCL7A-containing SWI/SNF/BAF complexes to mitochondria-driven NPC commitment, thereby providing a better understanding of the cell-intrinsic transcriptional processes that connect metabolism, neuronal morphogenesis, and cognitive flexibility.


Cell Differentiation , Microfilament Proteins , Neural Stem Cells , Animals , Mice , Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly , Energy Metabolism , Mitochondria/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Microfilament Proteins/metabolism , Neural Stem Cells/cytology
5.
Cell Death Differ ; 29(12): 2459-2471, 2022 12.
Article En | MEDLINE | ID: mdl-36138226

Oncohistones represent compelling evidence for a causative role of epigenetic perturbations in cancer. Giant cell tumours of bone (GCTs) are characterised by a mutated histone H3.3 as the sole genetic driver present in bone-forming osteoprogenitor cells but absent from abnormally large bone-resorbing osteoclasts which represent the hallmark of these neoplasms. While these striking features imply a pathogenic interaction between mesenchymal and myelomonocytic lineages during GCT development, the underlying mechanisms remain unknown. We show that the changes in the transcriptome and epigenome in the mesenchymal cells caused by the H3.3-G34W mutation contribute to increase osteoclast recruitment in part via reduced expression of the TGFß-like soluble factor, SCUBE3. Transcriptional changes in SCUBE3 are associated with altered histone marks and H3.3G34W enrichment at its enhancer regions. In turn, osteoclasts secrete unregulated amounts of SEMA4D which enhances proliferation of mutated osteoprogenitors arresting their maturation. These findings provide a mechanism by which GCTs undergo differentiation in response to denosumab, a drug that depletes the tumour of osteoclasts. In contrast, hTERT alterations, commonly found in malignant GCT, result in the histone-mutated neoplastic cells being independent of osteoclasts for their proliferation, predicting unresponsiveness to denosumab. We provide a mechanism for the initiation of GCT, the basis of which is dysfunctional cross-talk between bone-forming and bone-resorbing cells. The findings highlight the role of tumour/microenvironment bidirectional interactions in tumorigenesis and how this is exploited in the treatment of GCT.


Bone Neoplasms , Giant Cell Tumor of Bone , Humans , Giant Cell Tumor of Bone/genetics , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/pathology , Histones/genetics , Histones/metabolism , Denosumab/metabolism , Denosumab/therapeutic use , Bone Neoplasms/genetics , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Osteoclasts/metabolism , Bone Remodeling/genetics , Tumor Microenvironment , Calcium-Binding Proteins/metabolism
7.
Neuro Oncol ; 24(5): 741-754, 2022 05 04.
Article En | MEDLINE | ID: mdl-34865163

BACKGROUND: Developmental brain tumors harboring BRAFV600E somatic mutation are diverse. Here, we describe molecular factors that determine BRAFV600E-induced tumor biology and function. METHODS: Intraventricular in utero electroporation in combination with the piggyBac transposon system was utilized to generate developmental brain neoplasms, which were comprehensively analyzed with regard to growth using near-infrared in-vivo imaging, transcript signatures by RNA sequencing, and neuronal activity by multielectrode arrays. RESULTS: BRAF  V600E expression in murine neural progenitors elicits benign neoplasms composed of enlarged dysmorphic neurons and neoplastic astroglia recapitulating ganglioglioma (GG) only in concert with active Akt/mTOR-signaling. Purely glial tumors resembling aspects of polymorphous low-grade neuroepithelial tumors of the young (PLNTYs) emerge from BRAFV600E alone. Additional somatic Trp53-loss is sufficient to generate anaplastic GGs (aGGs) with glioneuronal clonality. Functionally, only BRAFV600E/pAkt tumors intrinsically generate substantial neuronal activity and show enhanced relay to adjacent tissue conferring high epilepsy propensity. In contrast, PLNTY- and aGG models lack significant spike activity, which appears in line with the glial differentiation of the former and a dysfunctional tissue structure combined with reduced neuronal transcript signatures in the latter. CONCLUSION: mTOR-signaling and Trp53-loss critically determine the biological diversity and electrical activity of BRAFV600E-induced tumors.


Brain Neoplasms , Ganglioglioma , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Ganglioglioma/genetics , Humans , Mice , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
8.
Nat Cell Biol ; 23(12): 1224-1239, 2021 12.
Article En | MEDLINE | ID: mdl-34876685

Defective silencing of retrotransposable elements has been linked to inflammageing, cancer and autoimmune diseases. However, the underlying mechanisms are only partially understood. Here we implicate the histone H3.3 chaperone Daxx, a retrotransposable element repressor inactivated in myeloid leukaemia and other neoplasms, in protection from inflammatory disease. Loss of Daxx alters the chromatin landscape, H3.3 distribution and histone marks of haematopoietic progenitors, leading to engagement of a Pu.1-dependent transcriptional programme for myelopoiesis at the expense of B-cell differentiation. This causes neutrophilia and inflammation, predisposing mice to develop an autoinflammatory skin disease. While these molecular and phenotypic perturbations are in part reverted in animals lacking both Pu.1 and Daxx, haematopoietic progenitors in these mice show unique chromatin and transcriptome alterations, suggesting an interaction between these two pathways. Overall, our findings implicate retrotransposable element silencing in haematopoiesis and suggest a cross-talk between the H3.3 loading machinery and the pioneer transcription factor Pu.1.


Chromatin/pathology , Co-Repressor Proteins/genetics , Leukocyte Disorders/congenital , Molecular Chaperones/genetics , Myelopoiesis/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , B-Lymphocytes/cytology , Cell Line , Chromatin/genetics , Hematopoietic Stem Cells/cytology , Histones/metabolism , Humans , Inflammation/pathology , Leukocyte Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Retroelements/genetics , Skin Diseases/genetics , Skin Diseases/immunology , Skin Diseases/pathology
10.
Curr Opin Pharmacol ; 60: 117-122, 2021 10.
Article En | MEDLINE | ID: mdl-34411982

With the aging of the population, Alzheimer's disease and other forms of dementia represent major challenges for health care systems globally. To date, the molecular mechanisms underlying the pathophysiology of dementia remain elusive, with a consequent negative impact in developing efficient disease modifiers. New exciting findings suggest that modulation of the histone code may influence transcriptional networks at the root of neuronal plasticity and cognitive performance. Although most of the current conclusions require further mechanistic evidence, it appears that chromatin perturbations actually correlate with Alzheimer's disease onset and progression. Thus, a better understanding of the epigenetic contribution to normal brain function and dementia pathogenesis may help to identify new epigenetic targets for the inhibition of disease trajectories associated with cognitive decline.


Alzheimer Disease , Cognitive Dysfunction , Aging/genetics , Alzheimer Disease/genetics , Chromatin/genetics , Histone Code , Humans
11.
Cell Death Dis ; 12(8): 785, 2021 08 11.
Article En | MEDLINE | ID: mdl-34381018

Pediatric gliomas comprise a broad range of brain tumors derived from glial cells. While high-grade gliomas are often resistant to therapy and associated with a poor outcome, children with low-grade gliomas face a better prognosis. However, the treatment of low-grade gliomas is often associated with severe long-term adverse effects. This shows that there is a strong need for improved treatment approaches. Here, we highlight the potential for repurposing disulfiram to treat pediatric gliomas. Disulfiram is a drug used to support the treatment of chronic alcoholism and was found to be effective against diverse cancer types in preclinical studies. Our results show that disulfiram efficiently kills pediatric glioma cell lines as well as patient-derived glioma stem cells. We propose a novel mechanism of action to explain disulfiram's anti-oncogenic activities by providing evidence that disulfiram induces the degradation of the oncoprotein MLL. Our results further reveal that disulfiram treatment and MLL downregulation induce similar responses at the level of histone modifications and gene expression, further strengthening that MLL is a key target of the drug and explaining its anti-oncogenic properties.


Alcoholism/drug therapy , Disulfiram/therapeutic use , Glioma/drug therapy , Glioma/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Proteolysis , Auranofin/pharmacology , Auranofin/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Disulfiram/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Repositioning , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/pathology , Histones/metabolism , Humans , Lysine/metabolism , Methylation/drug effects , Neoplasm Grading , Protein Processing, Post-Translational/drug effects , Proteolysis/drug effects , Transcription, Genetic/drug effects
12.
Neuro Oncol ; 23(11): 1885-1897, 2021 11 02.
Article En | MEDLINE | ID: mdl-33864086

BACKGROUND: Glioblastoma cells assemble to a syncytial communicating network based on tumor microtubes (TMs) as ultra-long membrane protrusions. The relationship between network architecture and transcriptional profile remains poorly investigated. Drugs that interfere with this syncytial connectivity such as meclofenamate (MFA) may be highly attractive for glioblastoma therapy. METHODS: In a human neocortical slice model using glioblastoma cell populations of different transcriptional signatures, three-dimensional tumor networks were reconstructed, and TM-based intercellular connectivity was mapped on the basis of two-photon imaging data. MFA was used to modulate morphological and functional connectivity; downstream effects of MFA treatment were investigated by RNA sequencing and fluorescence-activated cell sorting (FACS) analysis. RESULTS: TM-based network morphology strongly differed between the transcriptional cellular subtypes of glioblastoma and was dependent on axon guidance molecule expression. MFA revealed both a functional and morphological demolishment of glioblastoma network architectures which was reflected by a reduction of TM-mediated intercellular cytosolic traffic as well as a breakdown of TM length. RNA sequencing confirmed a downregulation of NCAM and axon guidance molecule signaling upon MFA treatment. Loss of glioblastoma communicating networks was accompanied by a failure in the upregulation of genes that are required for DNA repair in response to temozolomide (TMZ) treatment and culminated in profound treatment response to TMZ-mediated toxicity. CONCLUSION: The capacity of TM formation reflects transcriptional cellular heterogeneity. MFA effectively demolishes functional and morphological TM-based syncytial network architectures. These findings might pave the way to a clinical implementation of MFA as a TM-targeted therapeutic approach.


Brain Neoplasms , Glioblastoma , Meclofenamic Acid/pharmacology , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Glioblastoma/drug therapy , Humans , In Vitro Techniques
13.
J Clin Invest ; 131(1)2021 01 04.
Article En | MEDLINE | ID: mdl-33108356

Microglia maintain homeostasis in the brain. However, with age, they become primed and respond more strongly to inflammatory stimuli. We show here that microglia from aged mice had upregulated mTOR complex 1 signaling controlling translation, as well as protein levels of inflammatory mediators. Genetic ablation of mTOR signaling showed a dual yet contrasting effect on microglia priming: it caused an NF-κB-dependent upregulation of priming genes at the mRNA level; however, mice displayed reduced cytokine protein levels, diminished microglia activation, and milder sickness behavior. The effect on translation was dependent on reduced phosphorylation of 4EBP1, resulting in decreased binding of eIF4E to eIF4G. Similar changes were present in aged human microglia and in damage-associated microglia, indicating that upregulation of mTOR-dependent translation is an essential aspect of microglia priming in aging and neurodegeneration.


Aging/metabolism , Microglia/enzymology , Protein Biosynthesis , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Aging/genetics , Animals , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Humans , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation/genetics , TOR Serine-Threonine Kinases/genetics
15.
Cancer Cell ; 35(5): 782-797.e8, 2019 05 13.
Article En | MEDLINE | ID: mdl-31085178

High-grade gliomas defined by histone 3 K27M driver mutations exhibit global loss of H3K27 trimethylation and reciprocal gain of H3K27 acetylation, respectively shaping repressive and active chromatin landscapes. We generated tumor-derived isogenic models bearing this mutation and show that it leads to pervasive H3K27ac deposition across the genome. In turn, active enhancers and promoters are not created de novo and instead reflect the epigenomic landscape of the cell of origin. H3K27ac is enriched at repeat elements, resulting in their increased expression, which in turn can be further amplified by DNA demethylation and histone deacetylase inhibitors providing an exquisite therapeutic vulnerability. These agents may therefore modulate anti-tumor immune responses as a therapeutic modality for this untreatable disease.


Brain Neoplasms/metabolism , Glioma/metabolism , Histones/genetics , Histones/metabolism , Acetylation , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Chromatin/metabolism , Enhancer Elements, Genetic/drug effects , Epigenomics/methods , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Glioma/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Mutation
16.
Nat Commun ; 10(1): 1262, 2019 03 19.
Article En | MEDLINE | ID: mdl-30890717

Lys-27-Met mutations in histone 3 genes (H3K27M) characterize a subgroup of deadly gliomas and decrease genome-wide H3K27 trimethylation. Here we use primary H3K27M tumor lines and isogenic CRISPR-edited controls to assess H3K27M effects in vitro and in vivo. We find that whereas H3K27me3 and H3K27me2 are normally deposited by PRC2 across broad regions, their deposition is severely reduced in H3.3K27M cells. H3K27me3 is unable to spread from large unmethylated CpG islands, while H3K27me2 can be deposited outside these PRC2 high-affinity sites but to levels corresponding to H3K27me3 deposition in wild-type cells. Our findings indicate that PRC2 recruitment and propagation on chromatin are seemingly unaffected by K27M, which mostly impairs spread of the repressive marks it catalyzes, especially H3K27me3. Genome-wide loss of H3K27me3 and me2 deposition has limited transcriptomic consequences, preferentially affecting lowly-expressed genes regulating neurogenesis. Removal of H3K27M restores H3K27me2/me3 spread, impairs cell proliferation, and completely abolishes their capacity to form tumors in mice.


Brain Neoplasms/genetics , Chromatin/metabolism , Glioblastoma/genetics , Histones/genetics , Polycomb Repressive Complex 2/metabolism , Adolescent , Aged , Animals , Brain Neoplasms/pathology , CRISPR-Cas Systems , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Child , CpG Islands/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Female , Gene Editing/methods , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , HEK293 Cells , Histone Code/genetics , Histones/metabolism , Humans , Lysine/genetics , Male , Methionine/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neurogenesis/genetics , Xenograft Model Antitumor Assays
17.
Proc Natl Acad Sci U S A ; 116(14): 6975-6984, 2019 04 02.
Article En | MEDLINE | ID: mdl-30877245

Genomic instability (GI) drives tumor heterogeneity and promotes tumor progression and therapy resistance. However, causative factors underlying GI and means for clinical detection of GI in glioma are inadequately identified. We describe here that elevated expression of a gene module coexpressed with CDC20 (CDC20-M), the activator of the anaphase-promoting complex in the cell cycle, marks GI in glioma. The CDC20-M, containing 139 members involved in cell proliferation, DNA damage response, and chromosome segregation, was found to be consistently coexpressed in glioma transcriptomes. The coexpression of these genes was conserved across multiple species and organ systems, particularly in human neural stem and progenitor cells. CDC20-M expression was not correlated with the morphological subtypes, nor with the recently defined molecular subtypes of glioma. CDC20-M signature was an independent and robust predictor for poorer prognosis in over 1,000 patients from four large databases. Elevated CDC20-M signature enabled the identification of individual glioma samples with severe chromosome instability and mutation burden and of primary glioma cell lines with extensive mitotic errors leading to chromosome mis-segregation. AURKA, a core member of CDC20-M, was amplified in one-third of CDC20-M-high gliomas with gene-dosage-dependent expression. MLN8237, a Food and Drug Administration-approved AURKA inhibitor, selectively killed temozolomide-resistant primary glioma cells in vitro and prolonged the survival of a patient-derived xenograft mouse model with a high-CDC20-M signature. Our findings suggest that application of the CDC20-M signature may permit more selective use of adjuvant therapies for glioma patients and that dysregulated CDC20-M members may provide a therapeutic vulnerability in glioma.


Biomarkers, Tumor/biosynthesis , Cdc20 Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Genomic Instability , Glioma/metabolism , Neoplasm Proteins/biosynthesis , Animals , Biomarkers, Tumor/genetics , Cdc20 Proteins/genetics , Chemotherapy, Adjuvant , Female , Gene Expression Profiling , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/genetics , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
18.
Blood ; 131(6): 636-648, 2018 02 08.
Article En | MEDLINE | ID: mdl-29191918

A hallmark of acute promyelocytic leukemia (APL) is altered nuclear architecture, with disruption of promyelocytic leukemia (PML) nuclear bodies (NBs) mediated by the PML-retinoic acid receptor α (RARα) oncoprotein. To address whether this phenomenon plays a role in disease pathogenesis, we generated a knock-in mouse model with NB disruption mediated by 2 point mutations (C62A/C65A) in the Pml RING domain. Although no leukemias developed in PmlC62A/C65A mice, these transgenic mice also expressing RARα linked to a dimerization domain (p50-RARα model) exhibited a doubling in the rate of leukemia, with a reduced latency period. Additionally, we found that response to targeted therapy with all-trans retinoic acid in vivo was dependent on NB integrity. PML-RARα is recognized to be insufficient for development of APL, requiring acquisition of cooperating mutations. We therefore investigated whether NB disruption might be mutagenic. Compared with wild-type cells, primary PmlC62A/C65A cells exhibited increased sister-chromatid exchange and chromosome abnormalities. Moreover, functional assays showed impaired homologous recombination (HR) and nonhomologous end-joining (NHEJ) repair pathways, with defective localization of Brca1 and Rad51 to sites of DNA damage. These data directly demonstrate that Pml NBs are critical for DNA damage responses, and suggest that Pml NB disruption is a central contributor to APL pathogenesis.


DNA Repair/genetics , Intranuclear Inclusion Bodies/metabolism , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Promyelocytic Leukemia Protein/physiology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA Damage/genetics , DNA End-Joining Repair/genetics , Intranuclear Inclusion Bodies/genetics , Leukemia, Promyelocytic, Acute/metabolism , Mice , Mice, Transgenic , Mutagenesis/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promyelocytic Leukemia Protein/genetics , Signal Transduction/genetics
19.
J Natl Cancer Inst ; 110(5): 467-478, 2018 05 01.
Article En | MEDLINE | ID: mdl-29165716

Background: Imatinib and second-generation tyrosine kinase inhibitors (TKIs) nilotinib and dasatinib have statistically significantly improved the life expectancy of chronic myeloid leukemia (CML) patients; however, resistance to TKIs remains a major clinical challenge. Although ponatinib, a third-generation TKI, improves outcomes for patients with BCR-ABL-dependent mechanisms of resistance, including the T315I mutation, a proportion of patients may have or develop BCR-ABL-independent resistance and fail ponatinib treatment. By modeling ponatinib resistance and testing samples from these CML patients, it is hoped that an alternative drug target can be identified and inhibited with a novel compound. Methods: Two CML cell lines with acquired BCR-ABL-independent resistance were generated following culture in ponatinib. RNA sequencing and gene ontology (GO) enrichment were used to detect aberrant transcriptional response in ponatinib-resistant cells. A validated oncogene drug library was used to identify US Food and Drug Administration-approved drugs with activity against TKI-resistant cells. Validation was performed using bone marrow (BM)-derived cells from TKI-resistant patients (n = 4) and a human xenograft mouse model (n = 4-6 mice per group). All statistical tests were two-sided. Results: We show that ponatinib-resistant CML cells can acquire BCR-ABL-independent resistance mediated through alternative activation of mTOR. Following transcriptomic analysis and drug screening, we highlight mTOR inhibition as an alternative therapeutic approach in TKI-resistant CML cells. Additionally, we show that catalytic mTOR inhibitors induce autophagy and demonstrate that genetic or pharmacological inhibition of autophagy sensitizes ponatinib-resistant CML cells to death induced by mTOR inhibition in vitro (% number of colonies of control[SD], NVP-BEZ235 vs NVP-BEZ235+HCQ: 45.0[17.9]% vs 24.0[8.4]%, P = .002) and in vivo (median survival of NVP-BEZ235- vs NVP-BEZ235+HCQ-treated mice: 38.5 days vs 47.0 days, P = .04). Conclusion: Combined mTOR and autophagy inhibition may provide an attractive approach to target BCR-ABL-independent mechanism of resistance.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy/drug effects , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate/administration & dosage , Imidazoles/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Molecular Targeted Therapy/methods , Pyridazines/administration & dosage , Pyrimidines/administration & dosage , Quinolines/administration & dosage , Xenograft Model Antitumor Assays
20.
Sci Rep ; 7(1): 17055, 2017 12 06.
Article En | MEDLINE | ID: mdl-29213114

Chromatin remodelers have emerged as prominent regulators of epigenetic processes and potential drivers of various human pathologies. The multi-subunit chromatin-remodeling SWI/SNF complex determines gene expression programs and, consequently, contributes to the differentiation, maturation and plasticity of neurons. Here, we investigate the elusive biological function of Bcl7a and Bcl7b, two newly identified subunits of the SWI/SNF complex that are highly expressed throughout the brain. We generated ubiquitous and neuron-specific Bcl7a and Bcl7b single and double knockout mice. We provide evidence that Bcl7b is dispensable for animal survival as well as behavioral plasticity. Conversely, ubiquitous Bcl7a knockout results in perinatal lethality, while genetic deletion of Bcl7a in postmitotic neurons elicits motor abnormalities and affects dendritic branching of Purkinje cells, with no obvious synergistic relationship with Bcl7b. Collectively, our findings reveal novel insights into the cellular processes linked to BCL7-containing SWI/SNF complexes and their unrecognized roles in the brain.


Behavior, Animal/physiology , Microfilament Proteins/metabolism , Animals , Body Weight , Brain/metabolism , Cells, Cultured , Female , Locomotion/physiology , Male , Mice , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/metabolism , Purkinje Cells/cytology , Purkinje Cells/metabolism
...