Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 499
Filter
1.
Cell ; 184(18): 4753-4771.e27, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34388391

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by notorious resistance to current therapies attributed to inherent tumor heterogeneity and highly desmoplastic and immunosuppressive tumor microenvironment (TME). Unique proline isomerase Pin1 regulates multiple cancer pathways, but its role in the TME and cancer immunotherapy is unknown. Here, we find that Pin1 is overexpressed both in cancer cells and cancer-associated fibroblasts (CAFs) and correlates with poor survival in PDAC patients. Targeting Pin1 using clinically available drugs induces complete elimination or sustained remissions of aggressive PDAC by synergizing with anti-PD-1 and gemcitabine in diverse model systems. Mechanistically, Pin1 drives the desmoplastic and immunosuppressive TME by acting on CAFs and induces lysosomal degradation of the PD-1 ligand PD-L1 and the gemcitabine transporter ENT1 in cancer cells, besides activating multiple cancer pathways. Thus, Pin1 inhibition simultaneously blocks multiple cancer pathways, disrupts the desmoplastic and immunosuppressive TME, and upregulates PD-L1 and ENT1, rendering PDAC eradicable by immunochemotherapy.


Subject(s)
Immunotherapy , Molecular Targeted Therapy , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Allografts/immunology , Amino Acid Motifs , Animals , Apoptosis/drug effects , B7-H1 Antigen/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Synergism , Endocytosis/drug effects , Equilibrative Nucleoside Transporter 1/metabolism , Humans , Immunosuppression Therapy , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Oncogenes , Organoids/drug effects , Organoids/pathology , Signal Transduction/drug effects , Survival Analysis , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
2.
Cell ; 184(12): 3143-3162.e32, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34004147

ABSTRACT

Gene expression by RNA polymerase II (RNAPII) is tightly controlled by cyclin-dependent kinases (CDKs) at discrete checkpoints during the transcription cycle. The pausing checkpoint following transcription initiation is primarily controlled by CDK9. We discovered that CDK9-mediated, RNAPII-driven transcription is functionally opposed by a protein phosphatase 2A (PP2A) complex that is recruited to transcription sites by the Integrator complex subunit INTS6. PP2A dynamically antagonizes phosphorylation of key CDK9 substrates including DSIF and RNAPII-CTD. Loss of INTS6 results in resistance to tumor cell death mediated by CDK9 inhibition, decreased turnover of CDK9 phospho-substrates, and amplification of acute oncogenic transcriptional responses. Pharmacological PP2A activation synergizes with CDK9 inhibition to kill both leukemic and solid tumor cells, providing therapeutic benefit in vivo. These data demonstrate that fine control of gene expression relies on the balance between kinase and phosphatase activity throughout the transcription cycle, a process dysregulated in cancer that can be exploited therapeutically.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Protein Phosphatase 2/metabolism , RNA-Binding Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Phosphorylation , Protein Binding , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Substrate Specificity
3.
Cell ; 181(3): 702-715.e20, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32315619

ABSTRACT

Protein phosphatase 2A (PP2A) enzymes can suppress tumors, but they are often inactivated in human cancers overexpressing inhibitory proteins. Here, we identify a class of small-molecule iHAPs (improved heterocyclic activators of PP2A) that kill leukemia cells by allosterically assembling a specific heterotrimeric PP2A holoenzyme consisting of PPP2R1A (scaffold), PPP2R5E (B56ε, regulatory), and PPP2CA (catalytic) subunits. One compound, iHAP1, activates this complex but does not inhibit dopamine receptor D2, a mediator of neurologic toxicity induced by perphenazine and related neuroleptics. The PP2A complex activated by iHAP1 dephosphorylates the MYBL2 transcription factor on Ser241, causing irreversible arrest of leukemia and other cancer cells in prometaphase. In contrast, SMAPs, a separate class of compounds, activate PP2A holoenzymes containing a different regulatory subunit, do not dephosphorylate MYBL2, and arrest tumor cells in G1 phase. Our findings demonstrate that small molecules can serve as allosteric switches to activate distinct PP2A complexes with unique substrates.


Subject(s)
Protein Phosphatase 2/metabolism , Apoptosis , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Enzyme Activators/metabolism , G1 Phase , Humans , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , Phenothiazines/pharmacology , Phosphorylation , Protein Phosphatase 2/physiology , Protein Subunits/metabolism , Trans-Activators/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism
4.
Cell ; 180(5): 968-983.e24, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32109415

ABSTRACT

Mammalian tissues engage in specialized physiology that is regulated through reversible modification of protein cysteine residues by reactive oxygen species (ROS). ROS regulate a myriad of biological processes, but the protein targets of ROS modification that drive tissue-specific physiology in vivo are largely unknown. Here, we develop Oximouse, a comprehensive and quantitative mapping of the mouse cysteine redox proteome in vivo. We use Oximouse to establish several paradigms of physiological redox signaling. We define and validate cysteine redox networks within each tissue that are tissue selective and underlie tissue-specific biology. We describe a common mechanism for encoding cysteine redox sensitivity by electrostatic gating. Moreover, we comprehensively identify redox-modified disease networks that remodel in aged mice, establishing a systemic molecular basis for the long-standing proposed links between redox dysregulation and tissue aging. We provide the Oximouse compendium as a framework for understanding mechanisms of redox regulation in physiology and aging.


Subject(s)
Aging/genetics , Cysteine/genetics , Proteins/genetics , Proteome/genetics , Aging/metabolism , Aging/pathology , Animals , Cysteine/metabolism , Humans , Mice , Organ Specificity/genetics , Oxidation-Reduction , Oxidative Stress/genetics , Proteomics/methods , Reactive Oxygen Species , Signal Transduction/genetics
5.
Cell ; 183(6): 1714-1731.e10, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33275901

ABSTRACT

Targeted protein degradation (TPD) refers to the use of small molecules to induce ubiquitin-dependent degradation of proteins. TPD is of interest in drug development, as it can address previously inaccessible targets. However, degrader discovery and optimization remains an inefficient process due to a lack of understanding of the relative importance of the key molecular events required to induce target degradation. Here, we use chemo-proteomics to annotate the degradable kinome. Our expansive dataset provides chemical leads for ∼200 kinases and demonstrates that the current practice of starting from the highest potency binder is an ineffective method for discovering active compounds. We develop multitargeted degraders to answer fundamental questions about the ubiquitin proteasome system, uncovering that kinase degradation is p97 dependent. This work will not only fuel kinase degrader discovery, but also provides a blueprint for evaluating targeted degradation across entire gene families to accelerate understanding of TPD beyond the kinome.


Subject(s)
Protein Kinases/metabolism , Proteolysis , Proteome/metabolism , Adult , Cell Line , Databases, Protein , Female , Humans , Male , Middle Aged , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/genetics , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/metabolism , Young Adult
6.
Cell ; 174(2): 300-311.e11, 2018 07 12.
Article in English | MEDLINE | ID: mdl-30007416

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) recognition of cytosolic DNA is critical for immune responses to pathogen replication, cellular stress, and cancer. Existing structures of the mouse cGAS-DNA complex provide a model for enzyme activation but do not explain why human cGAS exhibits severely reduced levels of cyclic GMP-AMP (cGAMP) synthesis compared to other mammals. Here, we discover that enhanced DNA-length specificity restrains human cGAS activation. Using reconstitution of cGAMP signaling in bacteria, we mapped the determinant of human cGAS regulation to two amino acid substitutions in the DNA-binding surface. Human-specific substitutions are necessary and sufficient to direct preferential detection of long DNA. Crystal structures reveal why removal of human substitutions relaxes DNA-length specificity and explain how human-specific DNA interactions favor cGAS oligomerization. These results define how DNA-sensing in humans adapted for enhanced specificity and provide a model of the active human cGAS-DNA complex to enable structure-guided design of cGAS therapeutics.


Subject(s)
DNA/metabolism , Immunologic Surveillance/physiology , Nucleotidyltransferases/metabolism , Animals , Benzofurans/chemistry , Benzofurans/metabolism , Binding Sites , Catalytic Domain , Chemotaxis/drug effects , DNA/chemistry , Humans , Mice , Molecular Docking Simulation , Mutagenesis, Site-Directed , Nucleotides, Cyclic/metabolism , Nucleotides, Cyclic/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/genetics , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Species Specificity , Vibrio cholerae/metabolism , Vibrio cholerae/physiology
8.
Cell ; 171(7): 1573-1588.e28, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29224777

ABSTRACT

There is considerable evidence that chromosome structure plays important roles in gene control, but we have limited understanding of the proteins that contribute to structural interactions between gene promoters and their enhancer elements. Large DNA loops that encompass genes and their regulatory elements depend on CTCF-CTCF interactions, but most enhancer-promoter interactions do not employ this structural protein. Here, we show that the ubiquitously expressed transcription factor Yin Yang 1 (YY1) contributes to enhancer-promoter structural interactions in a manner analogous to DNA interactions mediated by CTCF. YY1 binds to active enhancers and promoter-proximal elements and forms dimers that facilitate the interaction of these DNA elements. Deletion of YY1 binding sites or depletion of YY1 protein disrupts enhancer-promoter looping and gene expression. We propose that YY1-mediated enhancer-promoter interactions are a general feature of mammalian gene control.


Subject(s)
Enhancer Elements, Genetic , Promoter Regions, Genetic , YY1 Transcription Factor/metabolism , Animals , CCCTC-Binding Factor/metabolism , Embryonic Stem Cells/metabolism , Humans , Mice
9.
Cell ; 171(6): 1437-1452.e17, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29195078

ABSTRACT

We previously piloted the concept of a Connectivity Map (CMap), whereby genes, drugs, and disease states are connected by virtue of common gene-expression signatures. Here, we report more than a 1,000-fold scale-up of the CMap as part of the NIH LINCS Consortium, made possible by a new, low-cost, high-throughput reduced representation expression profiling method that we term L1000. We show that L1000 is highly reproducible, comparable to RNA sequencing, and suitable for computational inference of the expression levels of 81% of non-measured transcripts. We further show that the expanded CMap can be used to discover mechanism of action of small molecules, functionally annotate genetic variants of disease genes, and inform clinical trials. The 1.3 million L1000 profiles described here, as well as tools for their analysis, are available at https://clue.io.


Subject(s)
Gene Expression Profiling/methods , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Profiling/economics , Humans , Neoplasms/drug therapy , Organ Specificity , Pharmaceutical Preparations/metabolism , Sequence Analysis, RNA/economics , Sequence Analysis, RNA/methods , Small Molecule Libraries
10.
Cell ; 166(3): 740-754, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27397505

ABSTRACT

Systematic studies of cancer genomes have provided unprecedented insights into the molecular nature of cancer. Using this information to guide the development and application of therapies in the clinic is challenging. Here, we report how cancer-driven alterations identified in 11,289 tumors from 29 tissues (integrating somatic mutations, copy number alterations, DNA methylation, and gene expression) can be mapped onto 1,001 molecularly annotated human cancer cell lines and correlated with sensitivity to 265 drugs. We find that cell lines faithfully recapitulate oncogenic alterations identified in tumors, find that many of these associate with drug sensitivity/resistance, and highlight the importance of tissue lineage in mediating drug response. Logic-based modeling uncovers combinations of alterations that sensitize to drugs, while machine learning demonstrates the relative importance of different data types in predicting drug response. Our analysis and datasets are rich resources to link genotypes with cellular phenotypes and to identify therapeutic options for selected cancer sub-populations.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Analysis of Variance , Cell Line, Tumor , DNA Methylation , Drug Resistance, Neoplasm/genetics , Gene Dosage , Humans , Models, Genetic , Mutation , Neoplasms/genetics , Oncogenes , Precision Medicine
11.
Cell ; 163(1): 174-86, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26406377

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer that exhibits extremely high levels of genetic complexity and yet a relatively uniform transcriptional program. We postulate that TNBC might be highly dependent on uninterrupted transcription of a key set of genes within this gene expression program and might therefore be exceptionally sensitive to inhibitors of transcription. Utilizing kinase inhibitors and CRISPR/Cas9-mediated gene editing, we show here that triple-negative but not hormone receptor-positive breast cancer cells are exceptionally dependent on CDK7, a transcriptional cyclin-dependent kinase. TNBC cells are unique in their dependence on this transcriptional CDK and suffer apoptotic cell death upon CDK7 inhibition. An "Achilles cluster" of TNBC-specific genes is especially sensitive to CDK7 inhibition and frequently associated with super-enhancers. We conclude that CDK7 mediates transcriptional addiction to a vital cluster of genes in TNBC and CDK7 inhibition may be a useful therapy for this challenging cancer.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , Transcription, Genetic , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Cyclin-Dependent Kinase-Activating Kinase
12.
Nature ; 628(8007): 442-449, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538798

ABSTRACT

Whereas oncogenes can potentially be inhibited with small molecules, the loss of tumour suppressors is more common and is problematic because the tumour-suppressor proteins are no longer present to be targeted. Notable examples include SMARCB1-mutant cancers, which are highly lethal malignancies driven by the inactivation of a subunit of SWI/SNF (also known as BAF) chromatin-remodelling complexes. Here, to generate mechanistic insights into the consequences of SMARCB1 mutation and to identify vulnerabilities, we contributed 14 SMARCB1-mutant cell lines to a near genome-wide CRISPR screen as part of the Cancer Dependency Map Project1-3. We report that the little-studied gene DDB1-CUL4-associated factor 5 (DCAF5) is required for the survival of SMARCB1-mutant cancers. We show that DCAF5 has a quality-control function for SWI/SNF complexes and promotes the degradation of incompletely assembled SWI/SNF complexes in the absence of SMARCB1. After depletion of DCAF5, SMARCB1-deficient SWI/SNF complexes reaccumulate, bind to target loci and restore SWI/SNF-mediated gene expression to levels that are sufficient to reverse the cancer state, including in vivo. Consequently, cancer results not from the loss of SMARCB1 function per se, but rather from DCAF5-mediated degradation of SWI/SNF complexes. These data indicate that therapeutic targeting of ubiquitin-mediated quality-control factors may effectively reverse the malignant state of some cancers driven by disruption of tumour suppressor complexes.


Subject(s)
Multiprotein Complexes , Mutation , Neoplasms , SMARCB1 Protein , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , CRISPR-Cas Systems , Gene Editing , Neoplasms/genetics , Neoplasms/metabolism , SMARCB1 Protein/deficiency , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Proteolysis , Ubiquitin/metabolism
13.
Cell ; 159(5): 1126-1139, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25416950

ABSTRACT

The MYC oncoproteins are thought to stimulate tumor cell growth and proliferation through amplification of gene transcription, a mechanism that has thwarted most efforts to inhibit MYC function as potential cancer therapy. Using a covalent inhibitor of cyclin-dependent kinase 7 (CDK7) to disrupt the transcription of amplified MYCN in neuroblastoma cells, we demonstrate downregulation of the oncoprotein with consequent massive suppression of MYCN-driven global transcriptional amplification. This response translated to significant tumor regression in a mouse model of high-risk neuroblastoma, without the introduction of systemic toxicity. The striking treatment selectivity of MYCN-overexpressing cells correlated with preferential downregulation of super-enhancer-associated genes, including MYCN and other known oncogenic drivers in neuroblastoma. These results indicate that CDK7 inhibition, by selectively targeting the mechanisms that promote global transcriptional amplification in tumor cells, may be useful therapy for cancers that are driven by MYC family oncoproteins.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Disease Models, Animal , Neuroblastoma/drug therapy , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Phenylenediamines/therapeutic use , Protein Kinase Inhibitors/pharmacology , Pyrimidines/therapeutic use , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Humans , N-Myc Proto-Oncogene Protein , Transcription, Genetic/drug effects , Cyclin-Dependent Kinase-Activating Kinase
14.
Nature ; 620(7973): 417-425, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37495688

ABSTRACT

Genes that drive the proliferation, survival, invasion and metastasis of malignant cells have been identified for many human cancers1-4. Independent studies have identified cell death pathways that eliminate cells for the good of the organism5,6. The coexistence of cell death pathways with driver mutations suggests that the cancer driver could be rewired to activate cell death using chemical inducers of proximity (CIPs). Here we describe a new class of molecules called transcriptional/epigenetic CIPs (TCIPs) that recruit the endogenous cancer driver, or a downstream transcription factor, to the promoters of cell death genes, thereby activating their expression. We focused on diffuse large B cell lymphoma, in which the transcription factor B cell lymphoma 6 (BCL6) is deregulated7. BCL6 binds to the promoters of cell death genes and epigenetically suppresses their expression8. We produced TCIPs by covalently linking small molecules that bind BCL6 to those that bind to transcriptional activators that contribute to the oncogenic program, such as BRD4. The most potent molecule, TCIP1, increases binding of BRD4 by 50% over genomic BCL6-binding sites to produce transcriptional elongation at pro-apoptotic target genes within 15 min, while reducing binding of BRD4 over enhancers by only 10%, reflecting a gain-of-function mechanism. TCIP1 kills diffuse large B cell lymphoma cell lines, including chemotherapy-resistant, TP53-mutant lines, at EC50 of 1-10 nM in 72 h and exhibits cell-specific and tissue-specific effects, capturing the combinatorial specificity inherent to transcription. The TCIP concept also has therapeutic applications in regulating the expression of genes for regenerative medicine and developmental disorders.


Subject(s)
Apoptosis , Gene Expression Regulation, Neoplastic , Lymphoma, Large B-Cell, Diffuse , Transcription Factors , Humans , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Transcription Factors/metabolism , Epigenesis, Genetic/drug effects , Promoter Regions, Genetic , Carcinogenesis/drug effects , Carcinogenesis/genetics
15.
Cell ; 145(1): 19-24, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21458664

ABSTRACT

Building on years of basic scientific discovery, recent advances in the fields of cancer genetics and medicinal chemistry are now converging to revolutionize the treatment of cancer. Starting with serendipitous observations in rare subsets of cancer, a paradigm shift in clinical research is poised to ensure that new molecular insights are rapidly applied to shape emerging cancer therapies. Could this mark a turning point in the "War on Cancer"?


Subject(s)
Biomedical Research/economics , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Drug Delivery Systems , Humans , Neoplasms/genetics
16.
Blood ; 141(23): 2841-2852, 2023 06 08.
Article in English | MEDLINE | ID: mdl-36877894

ABSTRACT

Therapeutic targeting of CDK7 has proven beneficial in preclinical studies, yet the off-target effects of currently available CDK7 inhibitors make it difficult to pinpoint the exact mechanisms behind MM cell death mediated by CDK7 inhibition. Here, we show that CDK7 expression positively correlates with E2F and MYC transcriptional programs in cells from patients with multiple myeloma (MM); its selective targeting counteracts E2F activity via perturbation of the cyclin-dependent kinases/Rb axis and impairs MYC-regulated metabolic gene signatures translating into defects in glycolysis and reduced levels of lactate production in MM cells. CDK7 inhibition using the covalent small-molecule inhibitor YKL-5-124 elicits a strong therapeutic response with minimal effects on normal cells, and causes in vivo tumor regression, increasing survival in several mouse models of MM including a genetically engineered mouse model of MYC-dependent MM. Through its role as a critical cofactor and regulator of MYC and E2F activity, CDK7 is therefore a master regulator of oncogenic cellular programs supporting MM growth and survival, and a valuable therapeutic target providing rationale for development of YKL-5-124 for clinical use.


Subject(s)
Cyclin-Dependent Kinase-Activating Kinase , Multiple Myeloma , Animals , Mice , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Multiple Myeloma/genetics
17.
Nat Chem Biol ; 19(7): 815-824, 2023 07.
Article in English | MEDLINE | ID: mdl-36823351

ABSTRACT

Creatine kinases (CKs) provide local ATP production in periods of elevated energetic demand, such as during rapid anabolism and growth. Thus, creatine energetics has emerged as a major metabolic liability in many rapidly proliferating cancers. Whether CKs can be targeted therapeutically is unknown because no potent or selective CK inhibitors have been developed. Here we leverage an active site cysteine present in all CK isoforms to develop a selective covalent inhibitor of creatine phosphagen energetics, CKi. Using deep chemoproteomics, we discover that CKi selectively engages the active site cysteine of CKs in cells. A co-crystal structure of CKi with creatine kinase B indicates active site inhibition that prevents bidirectional phosphotransfer. In cells, CKi and its analogs rapidly and selectively deplete creatine phosphate, and drive toxicity selectively in CK-dependent acute myeloid leukemia. Finally, we use CKi to uncover an essential role for CKs in the regulation of proinflammatory cytokine production in macrophages.


Subject(s)
Creatine Kinase , Creatine , Creatine Kinase/chemistry , Creatine Kinase/metabolism , Creatine/pharmacology , Cysteine , Phosphotransferases , Protein Isoforms
18.
Nature ; 572(7771): 676-680, 2019 08.
Article in English | MEDLINE | ID: mdl-31391581

ABSTRACT

The CCCTC-binding factor (CTCF), which anchors DNA loops that organize the genome into structural domains, has a central role in gene control by facilitating or constraining interactions between genes and their regulatory elements1,2. In cancer cells, the disruption of CTCF binding at specific loci by somatic mutation3,4 or DNA hypermethylation5 results in the loss of loop anchors and consequent activation of oncogenes. By contrast, the germ-cell-specific paralogue of CTCF, BORIS (brother of the regulator of imprinted sites, also known as CTCFL)6, is overexpressed in several cancers7-9, but its contributions to the malignant phenotype remain unclear. Here we show that aberrant upregulation of BORIS promotes chromatin interactions in ALK-mutated, MYCN-amplified neuroblastoma10 cells that develop resistance to ALK inhibition. These cells are reprogrammed to a distinct phenotypic state during the acquisition of resistance, a process defined by the initial loss of MYCN expression followed by subsequent overexpression of BORIS and a concomitant switch in cellular dependence from MYCN to BORIS. The resultant BORIS-regulated alterations in chromatin looping lead to the formation of super-enhancers that drive the ectopic expression of a subset of proneural transcription factors that ultimately define the resistance phenotype. These results identify a previously unrecognized role of BORIS-to promote regulatory chromatin interactions that support specific cancer phenotypes.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/genetics , Animals , CCCTC-Binding Factor/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Mice , Molecular Targeted Therapy , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/enzymology , Neuroblastoma/genetics , Phenotype , Protein Binding
20.
Mol Cell ; 67(1): 5-18.e19, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28673542

ABSTRACT

Processive elongation of RNA Polymerase II from a proximal promoter paused state is a rate-limiting event in human gene control. A small number of regulatory factors influence transcription elongation on a global scale. Prior research using small-molecule BET bromodomain inhibitors, such as JQ1, linked BRD4 to context-specific elongation at a limited number of genes associated with massive enhancer regions. Here, the mechanistic characterization of an optimized chemical degrader of BET bromodomain proteins, dBET6, led to the unexpected identification of BET proteins as master regulators of global transcription elongation. In contrast to the selective effect of bromodomain inhibition on transcription, BET degradation prompts a collapse of global elongation that phenocopies CDK9 inhibition. Notably, BRD4 loss does not directly affect CDK9 localization. These studies, performed in translational models of T cell leukemia, establish a mechanism-based rationale for the development of BET bromodomain degradation as cancer therapy.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Nuclear Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Transcription Elongation, Genetic , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/pharmacology , Cell Cycle Proteins , Cyclin-Dependent Kinase 9/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Leukemic , HCT116 Cells , HEK293 Cells , Humans , Jurkat Cells , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Multiprotein Complexes , Nuclear Proteins/genetics , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Stability , Proteolysis , RNA Polymerase II/metabolism , Time Factors , Transcription Elongation, Genetic/drug effects , Transcription Factors/genetics , Transfection , Ubiquitin-Protein Ligases , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL