143 results on '"John H. Bushweller"'
Search Results
2. Small Molecule Inhibitor of CBFβ-RUNX Binding for RUNX Transcription Factor Driven Cancers
- Author
-
Anuradha Illendula, Jane Gilmour, Jolanta Grembecka, Venkata Sesha Srimath Tirumala, Adam Boulton, Aravinda Kuntimaddi, Charles Schmidt, Lixin Wang, John A. Pulikkan, Hongliang Zong, Mahmut Parlak, Cem Kuscu, Anna Pickin, Yunpeng Zhou, Yan Gao, Lauren Mishra, Mazhar Adli, Lucio H. Castilla, Roger A. Rajewski, Kevin A. Janes, Monica L. Guzman, Constanze Bonifer, and John H. Bushweller
- Subjects
CBFβ ,RUNX ,PPI ,Transcription factor inhibitor ,Leukemia ,Triple negative breast cancer ,Medicine ,Medicine (General) ,R5-920 - Abstract
Transcription factors have traditionally been viewed with skepticism as viable drug targets, but they offer the potential for completely novel mechanisms of action that could more effectively address the stem cell like properties, such as self-renewal and chemo-resistance, that lead to the failure of traditional chemotherapy approaches. Core binding factor is a heterodimeric transcription factor comprised of one of 3 RUNX proteins (RUNX1-3) and a CBFβ binding partner. CBFβ enhances DNA binding of RUNX subunits by relieving auto-inhibition. Both RUNX1 and CBFβ are frequently mutated in human leukemia. More recently, RUNX proteins have been shown to be key players in epithelial cancers, suggesting the targeting of this pathway could have broad utility. In order to test this, we developed small molecules which bind to CBFβ and inhibit its binding to RUNX. Treatment with these inhibitors reduces binding of RUNX1 to target genes, alters the expression of RUNX1 target genes, and impacts cell survival and differentiation. These inhibitors show efficacy against leukemia cells as well as basal-like (triple-negative) breast cancer cells. These inhibitors provide effective tools to probe the utility of targeting RUNX transcription factor function in other cancers.
- Published
- 2016
- Full Text
- View/download PDF
3. Degree of Recruitment of DOT1L to MLL-AF9 Defines Level of H3K79 Di- and Tri-methylation on Target Genes and Transformation Potential
- Author
-
Aravinda Kuntimaddi, Nicholas J. Achille, Jeremy Thorpe, Alyson A. Lokken, Ritambhara Singh, Charles S. Hemenway, Mazhar Adli, Nancy J. Zeleznik-Le, and John H. Bushweller
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The MLL gene is a common target of chromosomal translocations found in human leukemia. MLL-fusion leukemia has a consistently poor outcome. One of the most common translocation partners is AF9 (MLLT3). MLL-AF9 recruits DOT1L, a histone 3 lysine 79 methyltransferase (H3K79me1/me2/me3), leading to aberrant gene transcription. We show that DOT1L has three AF9 binding sites and present the nuclear magnetic resonance (NMR) solution structure of a DOT1L-AF9 complex. We generate structure-guided point mutations and find that they have graded effects on recruitment of DOT1L to MLL-AF9. Chromatin immunoprecipitation sequencing (ChIP-seq) analyses of H3K79me2 and H3K79me3 show that graded reduction of the DOT1L interaction with MLL-AF9 results in differential loss of H3K79me2 and me3 at MLL-AF9 target genes. Furthermore, the degree of DOT1L recruitment is linked to the level of MLL-AF9 hematopoietic transformation.
- Published
- 2015
- Full Text
- View/download PDF
4. Bacillus anthracis Peptidoglycan Integrity Is Disrupted by the Chemokine CXCL10 through the FtsE/X Complex
- Author
-
Molly A. Hughes, Katie R. Margulieux, Benjamin K. Liebov, Venkata S. K. K. S. Tirumala, Arpita Singh, John H. Bushweller, and Robert K. Nakamoto
- Subjects
Bacillus anthracis ,chemokine ,CXCL10 ,antimicrobial ,FtsE/X ,peptidoglycan ,Microbiology ,QR1-502 - Abstract
The antimicrobial activity of the chemokine CXCL10 against vegetative cells of Bacillus anthracis occurs via both bacterial FtsE/X-dependent and-independent pathways. Previous studies established that the FtsE/X-dependent pathway was mediated through interaction of the N-terminal region(s) of CXCL10 with a functional FtsE/X complex, while the FtsE/X-independent pathway was mediated through the C-terminal α-helix of CXCL10. Both pathways result in cell lysis and death of B. anthracis. In other bacterial species, it has been shown that FtsE/X is involved in cellular elongation though activation of complex-associated peptidoglycan hydrolases. Thus, we hypothesized that the CXCL10-mediated killing of vegetative cells of B. anthracis through the FtsE/X-dependent pathway resulted from the disruption of peptidoglycan processing. Immunofluorescence microscopy studies using fluorescent peptidoglycan probes revealed that incubation of B. anthracis Sterne (parent) strain with CXCL10 or a C-terminal truncated CXCL10 (CTTC) affected peptidoglycan processing and/or incorporation of precursors into the cell wall. B. anthracis ΔftsX or ftsE(K123A/D481N) mutant strains, which lacked a functional FtsE/X complex, exhibited little to no evidence of disruption in peptidoglycan processing by either CXCL10 or CTTC. Additional studies demonstrated that the B. anthracis parent strain exhibited a statistically significant increase in peptidoglycan release in the presence of either CXCL10 or CTTC. While B. anthracis ΔftsX strain showed increased peptidoglycan release in the presence of CXCL10, no increase was observed with CTTC, suggesting that the FtsE/X-independent pathway was responsible for the activity observed with CXCL10. These results indicate that FtsE/X-dependent killing of vegetative cells of B. anthracis results from a loss of cell wall integrity due to disruption of peptidoglycan processing and suggest that FtsE/X may be an important antimicrobial target to study in the search for alternative microbial therapeutics.
- Published
- 2017
- Full Text
- View/download PDF
5. Corrigendum to: 'Small Molecule Inhibitor of CBFβ-RUNX Binding for RUNX Transcription Factor Driven Cancers' [EBioMedicine 8 (2016) 117–131]
- Author
-
Anuradha Illendula, Jane Gilmour, Jolanta Grembecka, Venkata Sesha Srimath Tirumala, Adam Boulton, Aravinda Kuntimaddi, Charles Schmidt, Lixin Wang, John A. Pulikkan, Hongliang Zong, Mahmut Parlak, Cem Kuscu, Anna Pickin, Yunpeng Zhou, Yan Gao, Lauren Mishra, Mazhar Adli, Lucio H. Castilla, Roger A. Rajewski, Kevin A. Janes, Monica L. Guzman, Constanze Bonifer, and John H. Bushweller
- Subjects
Medicine ,Medicine (General) ,R5-920 - Published
- 2017
- Full Text
- View/download PDF
6. Small-Molecule Inhibitors of the MLL1 CXXC Domain, an Epigenetic Reader of DNA Methylation
- Author
-
Hanuman P. Kalmode, Izabella Podsiadly, Ashish Kabra, Adam Boulton, Prabhakar Reddy, Yan Gao, Christopher Li, and John H. Bushweller
- Subjects
Organic Chemistry ,Drug Discovery ,Biochemistry - Abstract
The CXXC domain is a reader of DNA methylation which preferentially binds to unmethylated CpG DNA motifs. Chromosomal translocations involving the
- Published
- 2023
7. An erythroid-to-myeloid cell fate conversion is elicited by LSD1 inactivation
- Author
-
Sharon A. Singh, Yu Wang, Lei Yu, Takashi Moriguchi, Chia Jui Ku, Julien Pedron, Masayuki Yamamoto, Michael G. Rosenfeld, John H. Bushweller, Greggory Myers, Natee Jearawiriyapaisarn, James Douglas Engel, Rami Khoriaty, Andrew White, Kim-Chew Lim, and Emily Schneider
- Subjects
animal structures ,Myeloid ,Immunology ,Cell fate determination ,Biochemistry ,Cell Line ,Mice ,chemistry.chemical_compound ,Erythroid Cells ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Erythropoiesis ,Myeloid Cells ,Progenitor cell ,Transcription factor ,Cells, Cultured ,Histone Demethylases ,KDM1A ,Cell Biology ,Hematology ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,RUNX1 ,chemistry ,Gene Deletion - Abstract
Histone H3 lysine 4 methylation (H3K4Me) is most often associated with chromatin activation, and removing H3K4 methyl groups has been shown to be coincident with gene repression. H3K4Me demethylase KDM1a/LSD1 is a therapeutic target for multiple diseases, including for the potential treatment of β-globinopathies (sickle cell disease and β-thalassemia), because it is a component of γ-globin repressor complexes, and LSD1 inactivation leads to robust induction of the fetal globin genes. The effects of LSD1 inhibition in definitive erythropoiesis are not well characterized, so we examined the consequences of conditional inactivation of Lsd1 in adult red blood cells using a new Gata1creERT2 bacterial artificial chromosome transgene. Erythroid-specific loss of Lsd1 activity in mice led to a block in erythroid progenitor differentiation and to the expansion of granulocyte-monocyte progenitor–like cells, converting hematopoietic differentiation potential from an erythroid fate to a myeloid fate. The analogous phenotype was also observed in human hematopoietic stem and progenitor cells, coincident with the induction of myeloid transcription factors (eg, PU.1 and CEBPα). Finally, blocking the activity of the transcription factor PU.1 or RUNX1 at the same time as LSD1 inhibition rescued myeloid lineage conversion to an erythroid phenotype. These data show that LSD1 promotes erythropoiesis by repressing myeloid cell fate in adult erythroid progenitors and that inhibition of the myeloid-differentiation pathway reverses the lineage switch induced by LSD1 inactivation.
- Published
- 2021
- Full Text
- View/download PDF
8. The NOESY jigsaw: automated protein secondary structure and main-chain assignment from sparse, unassigned NMR data.
- Author
-
Christopher Bailey-Kellogg, Alik Widge, John J. Kelley 0001, Marcelo J. Berardi, John H. Bushweller, and Bruce Randall Donald
- Published
- 2000
- Full Text
- View/download PDF
9. Protein Disulfide Exchange by the Intramembrane Enzymes DsbB, DsbD, and CcdA
- Author
-
John H. Bushweller
- Subjects
Models, Molecular ,Cytochrome ,Protein Conformation ,medicine.disease_cause ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Bacterial Proteins ,Structural Biology ,Escherichia coli ,medicine ,Disulfides ,Molecular Biology ,Heme ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Escherichia coli Proteins ,Cytochrome c ,Cell Membrane ,Membrane Proteins ,Periplasmic space ,DsbA ,Membrane protein ,Biochemistry ,biology.protein ,Thioredoxin ,Oxidoreductases ,030217 neurology & neurosurgery - Abstract
The formation of disulfide bonds in proteins is an essential process in both prokaryotes and eukaryotes. In gram-negative bacteria including Escherichia coli, the proteins DsbA and DsbB mediate the formation of disulfide bonds in the periplasm. DsbA acts as the periplasmic oxidant of periplasmic substrate proteins. DsbA is reoxidized by transfer of reducing equivalents to the 4 TM helix membrane protein DsbB, which transfers reducing equivalents to ubiquinone or menaquinone. Multiple structural studies of DsbB have provided detailed structural information on intermediates in the process of DsbB catalyzed oxidation of DsbA. These structures and the insights gained are described. In proteins with more than one pair of Cys residues, there is the potential for formation of non-native disulfide bonds, making it necessary for the cell to have a mechanism for the isomerization of such non-native disulfide bonds. In E. coli, this is mediated by the proteins DsbC and DsbD. DsbC reduces mis-formed disulfide bonds. The eight-TM-helix protein DsbD reduces DsbC and is itself reduced by cytoplasmic thioredoxin. DsbD also contributes reducing equivalents for the reduction of cytochrome c to facilitate heme attachment. The DsbD functional homolog CcdA is a six-TM-helix membrane protein that provides reducing equivalents for the reduction of cytochrome c. A recent structure determination of CcdA has provided critical insights into how reducing equivalents are transferred across the membrane that likely also provides understanding how this is achieved by DsbD as well. This structure and the insights gained are described.
- Published
- 2020
- Full Text
- View/download PDF
10. Inhibition of the RUNX1-CBFβ transcription factor complex compromises mammary epithelial cell identity: a phenotype potentially stabilized by mitotic gene bookmarking
- Author
-
Jane B. Lian, Jonathan A. R. Gordon, Nicole A. Bouffard, John H. Bushweller, Andrew J. Fritz, Joseph R. Boyd, Sayyed K. Zaidi, Gary S. Stein, Janet L. Stein, Anuradha Illendula, Joshua T. Rose, and Eliana Moskovitz
- Subjects
0301 basic medicine ,RUNX1 ,Transcription factor complex ,Biology ,Cell morphology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,Gene expression ,Transcription factor ,Gene ,Mitosis ,CBFβ ,mitotic gene bookmarking ,RNA ,mammary epithelial phenotype ,Cell biology ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,embryonic structures ,epithelial phenotype ,Research Paper - Abstract
RUNX1 has recently been shown to play an important role in determination of mammary epithelial cell identity. However, mechanisms by which loss of the RUNX1 transcription factor in mammary epithelial cells leads to epithelial-to-mesenchymal transition (EMT) are not known. Here, we report that interaction between RUNX1 and its heterodimeric partner CBFβ is essential for sustaining mammary epithelial cell identity. Disruption of RUNX1-CBFβ interaction, DNA binding, and association with mitotic chromosomes alters cell morphology, global protein synthesis, and phenotype-related gene expression. During interphase, RUNX1 is organized as punctate, predominantly nuclear, foci that are dynamically redistributed during mitosis, with a subset localized to mitotic chromosomes. Genome-wide RUNX1 occupancy profiles for asynchronous, mitotically enriched, and early G1 breast epithelial cells reveal RUNX1 associates with RNA Pol II-transcribed protein coding and long non-coding RNA genes and RNA Pol I-transcribed ribosomal genes critical for mammary epithelial proliferation, growth, and phenotype maintenance. A subset of these genes remains occupied by the protein during the mitosis to G1 transition. Together, these findings establish that the RUNX1-CBFβ complex is required for maintenance of the normal mammary epithelial phenotype and its disruption leads to EMT. Importantly, our results suggest, for the first time, that RUNX1 mitotic bookmarking of a subset of epithelial-related genes may be an important epigenetic mechanism that contributes to stabilization of the mammary epithelial cell identity.
- Published
- 2020
11. RUNX1 and RUNX2 transcription factors function in opposing roles to regulate breast cancer stem cells
- Author
-
Jane B. Lian, Jason Kost, Joseph R. Boyd, Mark Fitzgerald, Seth Frietze, Deli Hong, Alqassem Abuarqoub, Prachi N. Ghule, Miles Malik, Janet L. Stein, John H. Bushweller, Sayyed K. Zaidi, Andrew J. Fritz, Jonathan A. R. Gordon, Sebastian Hanna, Kristiaan H. Finstaad, Gary S. Stein, and Coralee E. Tye
- Subjects
0301 basic medicine ,Epithelial-Mesenchymal Transition ,Physiology ,Clinical Biochemistry ,Breast Neoplasms ,Core Binding Factor Alpha 1 Subunit ,Mice, SCID ,Article ,CDH1 ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Biomarkers, Tumor ,Tumor Microenvironment ,Animals ,Humans ,RNA, Messenger ,Epithelial–mesenchymal transition ,skin and connective tissue diseases ,Transcription factor ,biology ,CD24 ,CD44 ,Cell Biology ,Gene Expression Regulation, Neoplastic ,RUNX2 ,030104 developmental biology ,RUNX1 ,chemistry ,030220 oncology & carcinogenesis ,Core Binding Factor Alpha 2 Subunit ,embryonic structures ,Neoplastic Stem Cells ,biology.protein ,Cancer research ,Heterografts ,Female ,Stem cell ,Signal Transduction - Abstract
Breast cancer stem cells (BCSCs) are competent to initiate tumor formation and growth and refractory to conventional therapies. Consequently BCSCs are implicated in tumor recurrence. Many signaling cascades associated with BCSCs are critical for epithelial-to-mesenchymal transition (EMT). We developed a model system to mechanistically examine BCSCs in basal-like breast cancer using MCF10AT1 FACS sorted for CD24 (negative/low in BCSCs) and CD44 (positive/high in BCSCs). Ingenuity Pathway Analysis comparing RNA-seq on the CD24-/low versus CD24+/high MCF10AT1 indicates that the top activated upstream regulators include TWIST1, TGFβ1, OCT4, and other factors known to be increased in BCSCs and during EMT. The top inhibited upstream regulators include ESR1, TP63, and FAS. Consistent with our results, many genes previously demonstrated to be regulated by RUNX factors are altered in BCSCs. The RUNX2 interaction network is the top significant pathway altered between CD24-/low and CD24+/high MCF10AT1. RUNX1 is higher in expression at the RNA level than RUNX2. RUNX3 is not expressed. While, human-specific quantitative polymerase chain reaction primers demonstrate that RUNX1 and CDH1 decrease in human MCF10CA1a cells that have grown tumors within the murine mammary fat pad microenvironment, RUNX2 and VIM increase. Treatment with an inhibitor of RUNX binding to CBFβ for 5 days followed by a 7-day recovery period results in EMT suggesting that loss of RUNX1, rather than increase in RUNX2, is a driver of EMT in early stage breast cancer. Increased understanding of RUNX regulation on BCSCs and EMT will provide novel insight into therapeutic strategies to prevent recurrence.
- Published
- 2020
- Full Text
- View/download PDF
12. Targeting transcription factors in cancer — from undruggable to reality
- Author
-
John H. Bushweller
- Subjects
Proteasome Endopeptidase Complex ,General Mathematics ,Antineoplastic Agents ,Ligands ,Article ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Transcription (biology) ,Catalytic Domain ,Cell Line, Tumor ,Neoplasms ,Protein Interaction Mapping ,Gene expression ,Gene duplication ,Animals ,Humans ,Phosphorylation ,Transcription factor ,Cell Proliferation ,Regulation of gene expression ,biology ,Kinase ,Applied Mathematics ,Point mutation ,Ubiquitination ,DNA ,Cell biology ,Gene Expression Regulation, Neoplastic ,Drug Design ,030220 oncology & carcinogenesis ,Mutation ,Proteolysis ,biology.protein ,Protein Processing, Post-Translational ,Protein Binding ,Transcription Factors - Abstract
Mutated or dysregulated transcription factors represent a unique class of drug targets that mediate aberrant gene expression, including blockade of differentiation and cell death gene expression programmes, hallmark properties of cancers. Transcription factor activity is altered in numerous cancer types via various direct mechanisms including chromosomal translocations, gene amplification or deletion, point mutations and alteration of expression, as well as indirectly through non-coding DNA mutations that affect transcription factor binding. Multiple approaches to target transcription factor activity have been demonstrated, preclinically and, in some cases, clinically, including inhibition of transcription factor–cofactor protein–protein interactions, inhibition of transcription factor–DNA binding and modulation of levels of transcription factor activity by altering levels of ubiquitylation and subsequent proteasome degradation or by inhibition of regulators of transcription factor expression. In addition, several new approaches to targeting transcription factors have recently emerged including modulation of auto-inhibition, proteolysis targeting chimaeras (PROTACs), use of cysteine reactive inhibitors, targeting intrinsically disordered regions of transcription factors and combinations of transcription factor inhibitors with kinase inhibitors to block the development of resistance. These innovations in drug development hold great promise to yield agents with unique properties that are likely to impact future cancer treatment.
- Published
- 2019
- Full Text
- View/download PDF
13. RUNX1-targeted therapy for AML expressing somatic or germline mutation in RUNX1
- Author
-
Yimin Qian, Warren Fiskus, Christopher P. Mill, David N. Saenz, John H. Bushweller, Tapan M. Kadia, Cristian Coarfa, Dyana T. Saenz, Kanak Raina, Andrew Futreal, Kimal Rajapakshe, Koichi Takahashi, Anuradha Illendula, Michael R. Green, Kapil N. Bhalla, Courtney D. DiNardo, Steven M. Kornblau, Craig M. Crews, Dimuthu Perera, and Joseph D. Khoury
- Subjects
Somatic cell ,Platelet disorder ,Immunology ,Antineoplastic Agents ,Apoptosis ,Biology ,Biochemistry ,Germline ,Small hairpin RNA ,Mice ,chemistry.chemical_compound ,Germline mutation ,Cell Line, Tumor ,hemic and lymphatic diseases ,Animals ,Humans ,Germ-Line Mutation ,Cell Proliferation ,Gene knockdown ,Myeloid Neoplasia ,Myeloid leukemia ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,Leukemia, Myeloid, Acute ,RUNX1 ,chemistry ,Gene Knockdown Techniques ,Core Binding Factor Alpha 2 Subunit ,embryonic structures ,Cancer research - Abstract
RUNX1 transcription factor regulates normal and malignant hematopoiesis. Somatic or germline mutant RUNX1 (mtRUNX1) is associated with poorer outcome in acute myeloid leukemia (AML). Knockdown or inhibition of RUNX1 induced more apoptosis of AML expressing mtRUNX1 versus wild-type RUNX1 and improved survival of mice engrafted with mtRUNX1-expressing AML. CRISPR/Cas9-mediated editing-out of RUNX1 enhancer (eR1) within its intragenic super-enhancer, or BET protein BRD4 depletion by short hairpin RNA, repressed RUNX1, inhibited cell growth, and induced cell lethality in AML cells expressing mtRUNX1. Moreover, treatment with BET protein inhibitor or degrader (BET–proteolysis targeting chimera) repressed RUNX1 and its targets, inducing apoptosis and improving survival of mice engrafted with AML expressing mtRUNX1. Library of Integrated Network–based Cellular Signatures 1000–connectivity mapping data sets queried with messenger RNA signature of RUNX1 knockdown identified novel expression-mimickers (EMs), which repressed RUNX1 and exerted in vitro and in vivo efficacy against AML cells expressing mtRUNX1. In addition, the EMs cinobufagin, anisomycin, and narciclasine induced more lethality in hematopoietic progenitor cells (HPCs) expressing germline mtRUNX1 from patients with AML compared with HPCs from patients with familial platelet disorder (FPD), or normal untransformed HPCs. These findings highlight novel therapeutic agents for AML expressing somatic or germline mtRUNX1.
- Published
- 2019
- Full Text
- View/download PDF
14. The intrinsically disordered proteins MLLT3 (AF9) and MLLT1 (ENL) – multimodal transcriptional switches with roles in normal hematopoiesis, MLL fusion leukemia, and kidney cancer
- Author
-
Ashish Kabra and John H. Bushweller
- Subjects
Population ,Intrinsically disordered proteins ,Wilms Tumor ,Chromatin remodeling ,Article ,03 medical and health sciences ,0302 clinical medicine ,Structural Biology ,hemic and lymphatic diseases ,Transcriptional regulation ,Humans ,Protein Interaction Domains and Motifs ,education ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,biology ,Chemistry ,Myeloid leukemia ,Nuclear Proteins ,DOT1L ,Kidney Neoplasms ,Bromodomain ,Cell biology ,Hematopoiesis ,Leukemia, Biphenotypic, Acute ,Neoplasm Proteins ,Intrinsically Disordered Proteins ,Histone ,Mutation ,biology.protein ,Gene Fusion ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
AF9 (MLLT3) and ENL (MLLT1) are members of the YEATS family (named after the five proteins first shown to contain this domain: Yaf9, ENL, AF9, Taf14, Sas5) defined by the presence of a YEATS domain. The YEATS domain is an epigenetic reader that binds to acetylated and crotonylated lysines, unlike the bromodomain which can only bind to acetylated lysines. All members of this family have been shown to be components of various complexes with roles in chromatin remodeling, histone modification, histone variant deposition, and transcriptional regulation. MLLT3 is a critical regulator of hematopoiesis with a role in maintaining the hematopoietic stem or progenitor cell (HSPC) population. Approximately 10% of acute myeloid leukemia (AML) and acute lymphocytic leukemia (ALL) patients harbor a translocation involving MLL (mixed lineage leukemia). In the context of MLL fusion patients with AML and ALL, MLL-AF9 and MLL-ENL fusions are observed in 34 and 31% of the patients, respectively. The intrinsically disordered C-terminal domain of MLLT3 (AHD, ANC1 homology domain) undergoes coupled binding and folding upon interaction with partner proteins AF4, DOT1L, BCOR, and CBX8. Backbone dynamics studies of the complexes suggest a role for dynamics in function. Inhibitors of the interaction of the intrinsically disordered AHD with partner proteins have been described, highlighting the feasibility of targeting intrinsically disordered regions. MLLT1 undergoes phase separation to enhance recruitment of the super elongation complex (SEC) and drive transcription. Mutations in MLLT1 observed in Wilms tumor patients enhance phase separation and transcription to drive an aberrant gene expression program.
- Published
- 2021
15. Regulome analysis in B-acute lymphoblastic leukemia exposes Core Binding Factor addiction as a therapeutic vulnerability
- Author
-
Jason P, Wray, Elitza M, Deltcheva, Charlotta, Boiers, Simon Е, Richardson, Jyoti Bikram, Chhetri, John, Brown, Sladjana, Gagrica, Yanping, Guo, Anuradha, Illendula, Joost H A, Martens, Hendrik G, Stunnenberg, John H, Bushweller, Rachael, Nimmo, and Tariq, Enver
- Subjects
Adult ,B-Lymphocytes ,Core Binding Factor Alpha 2 Subunit ,Core Binding Factors ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Gene Fusion ,Child - Abstract
The ETV6-RUNX1 onco-fusion arises in utero, initiating a clinically silent pre-leukemic state associated with the development of pediatric B-acute lymphoblastic leukemia (B-ALL). We characterize the ETV6-RUNX1 regulome by integrating chromatin immunoprecipitation- and RNA-sequencing and show that ETV6-RUNX1 functions primarily through competition for RUNX1 binding sites and transcriptional repression. In pre-leukemia, this results in ETV6-RUNX1 antagonization of cell cycle regulation by RUNX1 as evidenced by mass cytometry analysis of B-lineage cells derived from ETV6-RUNX1 knock-in human pluripotent stem cells. In frank leukemia, knockdown of RUNX1 or its co-factor CBFβ results in cell death suggesting sustained requirement for RUNX1 activity which is recapitulated by chemical perturbation using an allosteric CBFβ-inhibitor. Strikingly, we show that RUNX1 addiction extends to other genetic subtypes of pediatric B-ALL and also adult disease. Importantly, inhibition of RUNX1 activity spares normal hematopoiesis. Our results suggest that chemical intervention in the RUNX1 program may provide a therapeutic opportunity in ALL.
- Published
- 2021
16. Targeting Runt-Related Transcription Factor 1 Prevents Pulmonary Fibrosis and Reduces Expression of Severe Acute Respiratory Syndrome Coronavirus 2 Host Mediators
- Author
-
Leslie Ramos, Fabian Heinrich, Antonia Fitzek, Julien Pedron, Leo A. Kim, Natalia Chmielewska, Stefan Steurer, Xinyao Hu, Michael J. O'Hare, Benjamin Ondruschka, Santiago Delgado-Tirado, John H. Bushweller, Markus Glatzel, Susanne Krasemann, David Lagares, Hannah A.B. Whitmore, Diego Sepulveda-Falla, Anita Chandrahas, Claudia Marino, Dhanesh Amarnani, Paul P. Liu, Miranda An, and Joseph F. Arboleda-Velasquez
- Subjects
0301 basic medicine ,Male ,Pulmonary Fibrosis ,Inflammation ,Bleomycin ,Pathology and Forensic Medicine ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Fibrosis ,Pulmonary fibrosis ,medicine ,Animals ,030212 general & internal medicine ,Transcription factor ,Furin ,Lung ,Cells, Cultured ,biology ,business.industry ,COVID-19 ,Epithelial Cells ,Regular Article ,respiratory system ,medicine.disease ,respiratory tract diseases ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Treatment Outcome ,chemistry ,Core Binding Factor Alpha 2 Subunit ,biology.protein ,Cancer research ,Tumor necrosis factor alpha ,Female ,Angiotensin-Converting Enzyme 2 ,medicine.symptom ,business - Abstract
Pulmonary fibrosis (PF) can arise from unknown causes, as in idiopathic PF, or as a consequence of infections, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Current treatments for PF slow, but do not stop, disease progression. We report that treatment with a runt-related transcription factor 1 (RUNX1) inhibitor (Ro24-7429), previously found to be safe, although ineffective, as a Tat inhibitor in patients with HIV, robustly ameliorates lung fibrosis and inflammation in the bleomycin-induced PF mouse model. RUNX1 inhibition blunted fundamental mechanisms downstream pathologic mediators of fibrosis and inflammation, including transforming growth factor-β1 and tumor necrosis factor-α, in cultured lung epithelial cells, fibroblasts, and vascular endothelial cells, indicating pleiotropic effects. RUNX1 inhibition also reduced the expression of angiotensin-converting enzyme 2 and FES Upstream Region (FURIN), host proteins critical for SARS-CoV-2 infection, in mice and in vitro. A subset of human lungs with SARS-CoV-2 infection overexpress RUNX1. These data suggest that RUNX1 inhibition via repurposing of Ro24-7429 may be beneficial for PF and to battle SARS-CoV-2, by reducing expression of viral mediators and by preventing respiratory complications.
- Published
- 2021
17. The epigenetic eraser LSD1 lies at the apex of a reversible erythroid to myeloid cell fate decision
- Author
-
Emily Schneider, Andrew White, Yv Wang, Takashi Moriguchi, Kim-Chew Lim, James Douglas Engel, John H. Bushweller, Chia Jui Ku, Julien Pedron, Michael G. Rosenfeld, Natee Jearawiriyapaisarn, Sharon A. Singh, Lei Yu, Greggory Myers, Masayuki Yamamoto, and Rami Khoriaty
- Subjects
animal structures ,Myeloid ,Chemistry ,KDM1A ,Cell fate determination ,Cell biology ,chemistry.chemical_compound ,Haematopoiesis ,medicine.anatomical_structure ,RUNX1 ,hemic and lymphatic diseases ,CEBPA ,medicine ,Erythropoiesis ,Progenitor cell - Abstract
Histone H3 lysine 4 methylation (H3K4Me) is proximally associated with chromatin activation, and therefore removing H3K4 methyl groups is normally coincident with gene repression. H3K4Me demethylase KDM1a/LSD1 is a potential therapeutic target for multiple diseases, including for the treatment of the {beta}-globinopathies (sickle cell disease and {beta}-thalassemia) since it is a component of multiple{gamma} -globin repressor complexes, and its inactivation leads to robust induction of the fetal globin genes. However, the effects of LSD1 inhibition in definitive erythroid cells are not well characterized. Here we examined the consequences of erythroid-specific conditional inactivation of Lsd1 in vivo using a new Gata1creERT2 bacterial artificial chromosome (BAC) transgene. Conditional loss of Lsd1 in adult mice led to a differentiation block in erythroid progenitor cells and the surprising expansion of a GMP-like cell pool, apparently converting hematopoietic differentiation potential from an erythroid to a myeloid fate. The analogous phenotype was also observed in human cells: inactivation of LSD1 in hematopoietic stem and progenitor cells (HSPC) also blocked erythroid differentiation, coincident with robust induction of myeloid transcription factor genes (e.g. Pu.1 and Cebpa). Remarkably, blocking the activity of PU.1 or RUNX1 (a transcriptional activator of Pu.1) at the same time as blocking LSD1 activity reverted the myeloid lineage conversion back to an erythroid phenotype. Taken together, the data show that LSD1 maintains erythropoiesis by reversibly repressing a myeloid cell fate in adult erythroid cell precursors, and that inhibition of the myeloid differentiation pathway can reverse the negative effects of LSD1 inactivation on erythroid differentiation.
- Published
- 2021
- Full Text
- View/download PDF
18. Cell cycle corruption in a pre-leukemic ETV6-RUNX1 model exposes RUNX1 addiction as a therapeutic target in acute lymphoblastic leukemia
- Author
-
Sladjana Gagrica, Simon Richardson, Jason Wray, Jyoti Bikram Chettri, Joost H.A. Martens, Anuradha Illendula, John H. Bushweller, Elitza Deltcheva, Yan-Ping Guo, Tariq Enver, Hendrik G. Stunnenberg, Rachael Nimmo, and Charlotta Böiers
- Subjects
Programmed cell death ,Preleukemia ,Allosteric regulation ,Regulome ,Cell cycle ,Biology ,medicine.disease ,Leukemia ,chemistry.chemical_compound ,RUNX1 ,chemistry ,hemic and lymphatic diseases ,embryonic structures ,medicine ,Cancer research ,Induced pluripotent stem cell - Abstract
SummaryThe ETV6-RUNX1 onco-fusion arises in utero, initiating a clinically silent pre-leukemic state associated with the development of pediatric B-acute lymphoblastic leukemia (B-ALL). We characterize the ETV6-RUNX1 regulome by integrating chromatin immunoprecipitation- and RNA-sequencing and show that ETV6-RUNX1 functions primarily through competition for RUNX1 binding sites and transcriptional repression. In pre-leukemia, this results in ETV6-RUNX1 antagonization of cell cycle regulation by RUNX1 as evidenced by mass cytometry analysis of B-lineage cells derived from ETV6-RUNX1 knock-in human pluripotent stem cells. In frank leukemia, knockdown of RUNX1 or its co-factor CBFβ results in cell death suggesting sustained requirement for RUNX1 activity which is recapitulated by chemical perturbation using an allosteric CBFβ-inhibitor. Strikingly, we show that RUNX1 addiction extends to other genetic subtypes of pediatric B-ALL and also adult disease. Importantly, inhibition of RUNX1 activity spares normal hematopoiesis. Our results implicate chemical intervention in the RUNX1 program as an exciting therapeutic opportunity in ALL.
- Published
- 2020
- Full Text
- View/download PDF
19. TNF‐α signaling regulates RUNX1 function in endothelial cells
- Author
-
Dhanesh Amarnani, Miranda An, Michael J. O'Hare, John H. Bushweller, Joseph F. Arboleda-Velasquez, Lucia Gonzalez-Buendia, Julien Pedron, Hannah A.B. Whitmore, Santiago Delgado-Tirado, and Leo A. Kim
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,signaling pathway ,Vascular Endothelial Growth Factor A ,RUNX1 ,Angiogenesis ,MAP Kinase Signaling System ,p38 mitogen-activated protein kinases ,Stimulation ,Biochemistry ,Retina ,03 medical and health sciences ,angiogenesis ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,hemic and lymphatic diseases ,Genetics ,Animals ,Humans ,Retinopathy of Prematurity ,Molecular Biology ,Research Articles ,Cells, Cultured ,Diabetic Retinopathy ,Chemistry ,Tumor Necrosis Factor-alpha ,Endothelial Cells ,eye diseases ,Choroidal Neovascularization ,Up-Regulation ,Endothelial stem cell ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,TNF‐α ,Glucose ,embryonic structures ,Core Binding Factor Alpha 2 Subunit ,Cancer research ,endothelial cell ,Wet Macular Degeneration ,Tumor necrosis factor alpha ,Signal transduction ,030217 neurology & neurosurgery ,Biotechnology ,Research Article - Abstract
Runt‐related transcription factor 1 (RUNX1) acts as a mediator of aberrant retinal angiogenesis and has been implicated in the progression of proliferative diabetic retinopathy (PDR). Patients with PDR, retinopathy of prematurity (ROP), and wet age‐related macular degeneration (wet AMD) have been found to have elevated levels of Tumor Necrosis Factor‐alpha (TNF‐α) in the eye. In fibrovascular membranes (FVMs) taken from patients with PDR RUNX1 expression was increased in the vasculature, while in human retinal microvascular endothelial cells (HRMECs), TNF‐α stimulation causes increased RUNX1 expression, which can be modulated by RUNX1 inhibitors. Using TNF‐α pathway inhibitors, we determined that in HRMECs, TNF‐α‐induced RUNX1 expression occurs via JNK activation, while NF‐κB and p38/MAPK inhibition did not affect RUNX1 expression. JNK inhibitors were also effective at stopping high d‐glucose‐stimulated RUNX1 expression. We further linked JNK to RUNX1 through Activator Protein 1 (AP‐1) and investigated the JNK‐AP‐1‐RUNX1 regulatory feedback loop, which can be modulated by VEGF. Additionally, stimulation with TNF‐α and d‐glucose had an additive effect on RUNX1 expression, which was downregulated by VEGF modulation. These data suggest that the downregulation of RUNX1 in conjunction with anti‐VEGF agents may be important in future treatments for the management of diseases of pathologic ocular angiogenesis.
- Published
- 2020
20. BCOR Binding to MLL-AF9 Is Essential for Leukemia via Altered EYA1, SIX, and MYC Activity
- Author
-
Nancy J. Zeleznik-Le, Shubin Zhang, John H. Bushweller, Aravinda Kuntimaddi, Benjamin I. Leach, Adam Boulton, Charles Schmidt, and Nicholas J. Achille
- Subjects
Oncogene Proteins, Fusion ,Population ,Mutant ,Translocation, Genetic ,Article ,Mice ,hemic and lymphatic diseases ,Gene expression ,medicine ,Animals ,Progenitor cell ,education ,Gene ,neoplasms ,Cell Proliferation ,education.field_of_study ,Leukemia ,Chemistry ,Point mutation ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,General Medicine ,DOT1L ,medicine.disease ,Cell biology ,Repressor Proteins ,Protein Tyrosine Phosphatases ,Myeloid-Lymphoid Leukemia Protein - Abstract
MLL is a target of chromosomal translocations in acute leukemias with poor prognosis. The common MLL fusion partner AF9 (MLLT3) can directly bind to AF4, DOT1L, BCOR, and CBX8. To delineate the relevance of BCOR and CBX8 binding to MLL-AF9 for leukemogenesis, here we determine protein structures of AF9 complexes with CBX8 and BCOR, and show that binding of all four partners to AF9 is mutually exclusive. Using the structural analyses, we identify point mutations that selectively disrupt AF9 interactions with BCOR and CBX8. In bone marrow stem/progenitor cells expressing point mutant CBX8 or point mutant MLL-AF9, we show that disruption of direct CBX8/MLL-AF9 binding does not impact in vitro cell proliferation, whereas loss of direct BCOR/MLL-AF9 binding causes partial differentiation and increased proliferation. Strikingly, loss of MLL-AF9/BCOR binding abrogated its leukemogenic potential in a mouse model. The MLL-AF9 mutant deficient for BCOR binding reduces the expression of the EYA1 phosphatase and the protein level of c-Myc. Reduction in BCOR binding to MLL-AF9 alters a MYC-driven gene expression program, as well as altering expression of SIX-regulated genes, likely contributing to the observed reduction in the leukemia-initiating cell population. Significance: Direct recruitment of BCOR to MLL-AF9 is essential for leukemia via EYA1 phosphatase regulation, altering MYC and SIX gene expression programs. Specific partner binding (AF4, DOT1L, and BCOR) contributes in distinct ways to MLL leukemia. This may provide a rationale for combination DOT1L and EYA1 inhibition for MLL fusion leukemia treatment. This article is highlighted in the In This Issue feature, p. 127
- Published
- 2020
21. Structure of the Complex of an Iminopyridinedione Protein Tyrosine Phosphatase 4A3 Phosphatase Inhibitor with Human Serum Albumin
- Author
-
Isabella K. Blanco, John H. Bushweller, Elizabeth R. Sharlow, Peter Wipf, Kelley E. McQueeney, Ettore J. Rastelli, Mateusz P. Czub, Adam Boulton, Wladek Minor, Taber S. Maskrey, Nikhil R. Tasker, and John S. Lazo
- Subjects
0301 basic medicine ,Cell Survival ,Pyridines ,Allosteric regulation ,Phosphatase ,Serum albumin ,Serum Albumin, Human ,Protein tyrosine phosphatase ,Calorimetry ,Crystallography, X-Ray ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Binding site ,Enzyme Assays ,Pharmacology ,Binding Sites ,biology ,Chemistry ,Cell growth ,Albumin ,Correction ,Human serum albumin ,Molecular biology ,Neoplasm Proteins ,030104 developmental biology ,biology.protein ,Molecular Medicine ,Imines ,Protein Tyrosine Phosphatases ,030217 neurology & neurosurgery ,medicine.drug ,Half-Life - Abstract
Protein tyrosine phosphatase (PTP) 4A3 is frequently overexpressed in human solid tumors and hematologic malignancies and is associated with tumor cell invasion, metastasis, and a poor patient prognosis. Several potent, selective, and allosteric small molecule inhibitors of PTP4A3 were recently identified. A lead compound in the series, JMS-053 (7-imino-2-phenylthieno[3,2-c]pyridine-4,6(5H,7H)-dione), has a long plasma half-life (∼ 24 hours) in mice, suggesting possible binding to serum components. We confirmed by isothermal titration calorimetry that JMS-053 binds to human serum albumin. A single JMS-053 binding site was identified by X-ray crystallography in human serum albumin at drug site 3, which is also known as subdomain IB. The binding of JMS-053 to human serum albumin, however, did not markedly alter the overall albumin structure. In the presence of serum albumin, the potency of JMS-053 as an in vitro inhibitor of PTP4A3 and human A2780 ovarian cancer cell growth was reduced. The reversible binding of JMS-053 to serum albumin may serve to increase JMS-053’s plasma half-life and thus extend the delivery of the compound to tumors. SIGNIFICANCE STATEMENT X-ray crystallography revealed that a potent, reversible, first-in-class small molecule inhibitor of the oncogenic phosphatase protein tyrosine phosphatase 4A3 binds to at least one site on human serum albumin, which is likely to extend the compound’s plasma half-life and thus assist in drug delivery into tumors.
- Published
- 2020
22. The interaction between RUNX2 and core binding factor beta as a potential therapeutic target in canine osteosarcoma
- Author
-
Luke Anthony Wittenburg, Dayn Romero Godinez, Anuradha Illendula, Fernando Alegre, John H. Bushweller, and Amanda R. Ormonde
- Subjects
Core Binding Factor beta Subunit ,Cell ,Gene Expression ,Core Binding Factor Alpha 1 Subunit ,Core binding factor ,0403 veterinary science ,0302 clinical medicine ,Dog Diseases ,transcription factor ,Cancer ,Pediatric ,Osteosarcoma ,Tumor ,Chemistry ,04 agricultural and veterinary sciences ,RUNX2 ,medicine.anatomical_structure ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Combination ,Drug Therapy, Combination ,Development of treatments and therapeutic interventions ,Pediatric Research Initiative ,040301 veterinary sciences ,Cell Survival ,Pediatric Cancer ,Allosteric regulation ,Bone Neoplasms ,Antineoplastic Agents ,Canine Osteosarcoma ,Article ,Cell Line ,03 medical and health sciences ,Dogs ,Rare Diseases ,Drug Therapy ,Cell Line, Tumor ,osteosarcoma ,medicine ,core-binding factor beta ,Animals ,Veterinary Sciences ,Transcription factor ,novel therapeutic targets ,General Veterinary ,canine sarcoma ,medicine.disease ,Orphan Drug ,Cancer research - Abstract
Osteosarcoma remains the most common primary bone tumour in dogs with half of affected dogs unable to survive 1 year beyond diagnosis. New therapeutic options are needed to improve outcomes for this disease. Recent investigations into potential therapeutic targets have focused on cell surface molecules with little clear therapeutic benefit. Transcription factors and protein interactions represent underdeveloped areas of therapeutic drug development. We have utilized allosteric inhibitors of the core binding factor transcriptional complex, comprised of core binding factor beta (CBFβ) and RUNX2, in four canine osteosarcoma cell lines Active inhibitor compounds demonstrate anti-tumour activities with concentrations demonstrated to be achievable in vivo while an inactive, structural analogue has no activity. We show that CBFβ inhibitors are capable of inducing apoptosis, inhibiting clonogenic cell growth, altering cell cycle progression and impeding migration and invasion in a cell line-dependent manner. These effects coincide with a reduced interaction between RUNX2 and CBFβ and alterations in expression of RUNX2 target genes. We also show that addition of CBFβ inhibitors to the commonly used cytotoxic chemotherapeutic drugs doxorubicin and carboplatin leads to additive and/or synergistic anti-proliferative effects in canine osteosarcoma cell lines. Taken together, we have identified the interaction between components of the core binding factor transcriptional complex, RUNX2 and CBFβ, as a potential novel therapeutic target in canine osteosarcoma and provide justification for further investigations into the anti-tumour activities we describe here.
- Published
- 2020
23. Small molecule inhibition of the CBFβ/RUNX interaction decreases ovarian cancer growth and migration through alterations in genes related to epithelial-to-mesenchymal transition
- Author
-
John H. Bushweller, Yan Gao, Roger A. Rajewski, Anant Shah, Adam Boulton, Anne L. Carlton, Anuradha Illendula, David Wotton, Danielle C. Llaneza, and Charles N. Landen
- Subjects
0301 basic medicine ,Epithelial-Mesenchymal Transition ,Mice, Nude ,Cell Growth Processes ,Carcinoma, Ovarian Epithelial ,Article ,Small Molecule Libraries ,Mice ,Random Allocation ,03 medical and health sciences ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasms, Glandular and Epithelial ,Epithelial–mesenchymal transition ,Ovarian Neoplasms ,Gene knockdown ,business.industry ,Cell growth ,DNA replication ,Core Binding Factor alpha Subunits ,Obstetrics and Gynecology ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,Cell culture ,Cancer research ,Female ,Ovarian cancer ,business ,Wound healing - Abstract
Objective Ovarian cancer survival and treatment have improved minimally in the past 20years. Novel treatment strategies are needed to combat this disease. This study investigates the effects of chemical inhibition of the CBFβ/RUNX protein-protein interaction on ovarian cancer cell lines. Methods Ovarian cancer cell lines were treated with CBFβ/RUNX inhibitors, and the effects on proliferation, DNA replication, wound healing, and anchorage-independent growth were measured. RNA-Seq was performed on compound-treated cells to identify differentially expressed genes. Genes altered by compound treatment were targeted with siRNAs, and effects on DNA replication and wound healing were measured. Results Chemical inhibition of the CBFβ/RUNX interaction decreases ovarian cancer cell proliferation. Inhibitor treatment leads to an S-phase cell cycle delay, as indicated by an increased percentage of cells in S-phase, and a decreased DNA replication rate. Inhibitor treatment also reduces wound healing and anchorage-independent growth. RNA-Seq on compound-treated cells revealed changes in a small number of genes related to proliferation and epithelial-to-mesenchymal transition. siRNA-mediated knockdown of INHBA and MMP1 – two genes whose expression decreases with compound treatment – slowed DNA replication and impaired wound healing. Conclusions Chemical inhibition of the CBFβ/RUNX interaction is a viable strategy for the treatment of ovarian cancer.
- Published
- 2018
- Full Text
- View/download PDF
24. Solution structure and elevator mechanism of the membrane electron transporter CcdA
- Author
-
John H. Bushweller and Yunpeng Zhou
- Subjects
Models, Molecular ,0301 basic medicine ,Magnetic Resonance Spectroscopy ,030106 microbiology ,Plasma protein binding ,Article ,Protein Structure, Secondary ,03 medical and health sciences ,Protein structure ,Bacterial Proteins ,Structural Biology ,Oxidoreductase ,Cysteine ,Molecular Biology ,chemistry.chemical_classification ,biology ,Chemistry ,Thermus thermophilus ,Cell Membrane ,Membrane Transport Proteins ,biology.organism_classification ,Major facilitator superfamily ,Transport protein ,Protein Transport ,Transmembrane domain ,Cross-Linking Reagents ,030104 developmental biology ,Cytoplasm ,Archaeoglobus fulgidus ,Solvents ,Biophysics ,Oxidoreductases ,Oxidation-Reduction ,Protein Binding - Abstract
Membrane oxidoreductase CcdA plays a central role in supplying reducing equivalents from the bacterial cytoplasm to the envelope. It transports electrons across the membrane using a single pair of cysteines by a mechanism that has not yet been elucidated. Here we report an NMR structure of the Thermus thermophilus CcdA (TtCcdA) in an oxidized and outward-facing state. CcdA consists of two inverted structural repeats of three transmembrane helices (2 × 3-TM). We computationally modeled and experimentally validated an inward-facing state, which suggests that CcdA uses an elevator-type movement to shuttle the reactive cysteines across the membrane. CcdA belongs to the LysE superfamily, and thus its structure may be relevant to other LysE clan transporters. Structure comparisons of CcdA, semiSWEET, Pnu, and major facilitator superfamily (MFS) transporters provide insights into membrane transporter architecture and mechanism.
- Published
- 2018
- Full Text
- View/download PDF
25. The CBFβ-SMMHC/NRP1 Axis Regulates FLT3 and TGF-Beta Pathways in Inv(16) Acute Myeloid Leukemia
- Author
-
Rui Li, Scot A. Wolfe, Anneliese Carrascoso, John H. Bushweller, Mohd Hafiz Ahmad, Julie Zhu, Ruud Delwel, Mahesh Hegde, Monica L. Guzman, Lucio H. Castilla, and Roger Mulet-Lazaro
- Subjects
hemic and lymphatic diseases ,Immunology ,Neuropilin 1 ,TGF beta signaling pathway ,Cancer research ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,Biochemistry - Abstract
Signal transduction pathways regulate the proliferation and viability of acute myeloid leukemia (AML) blasts. The regulation in the expression of cytokine receptors in AML is not well understood. In this study, we investigated how the CBFβ-SMMHC fusion protein regulates expression of cytokine receptors in inv(16) AML, with focus on the co-receptor Neuropilin-1 (NRP1). Knock-down of CBFβ-SMMHC expression, utilizing shRNA transduction, induced G1 phase of cell cycle arrest and reduced the viability of inv(16) ME-1 cells in culture. Expression profile analysis of CBFβ-SMMHC knock-down cells revealed a significant repression of genes associated with transmembrane receptor protein kinase pathways, including NRP1 (-5 fold), FGFR1 (-4.2 fold) , FLT3 (-2 fold) and TGFBR2 (-1.2 fold). The expression of NRP1 was significantly upregulated in inv(16) AML cases when compared to other AML sub-types and to hematopoietic stem and progenitor cells. Functionally, NRP1 knock-down reduced the viability of ME-1 cells with a similar dynamics as when using CBFβ-SMMHC shRNAs. In addition, the proliferation of inv(16) AML cells was reduced 4.1-fold when treated with a function-blocking antibody for the FV/VIII extracellular NRP1 domain, while having no effect in non-inv(16) AML cells or when using blocking antibody for the CUB extracellular domain. Furthermore, deletion of Nrp1 by gene editing reduced the colony-forming unit capacity of primary mouse Cbfb +/MYH11 leukemic cells and extended the median leukemia latency in vivo. To identify the genes regulated by NRP1 in inv(16) AML, we analysed the transcription profile of NRP1 knock-down in ME-1 cells. Gene Set Enrichment and Pathway Analysis revealed a repression in STAT5 pathway, and in signalling receptor activity, including FLT3 (-1.8 fold) and TGFBR2 (-1.8 fold) expression, indicating that NRP1 mediates transcriptional regulation of FLT3 and TGFBR2 expression in inv(16) AML. Furthermore, the regulation of FLT3 and TGFB2 expression by CBFβ-SMMHC and by NRP1 was validated by gene editing in inv(16) AML blasts. Accordingly, NRP1 knock-down in AML cells reduced SMAD2/3 phosphorylation. The repression of RUNX1/CBFβ function, using small molecule inhibitors, in inv(16) AML cells with CBFβ-SMMHC knockdown restored NRP1 expression, suggesting that RUNX1 may repress NRP1 expression in AML cells. To evaluate if RUNX1 directly regulates NRP1 expression, we tested RUNX1 binding in the NRP1 locus of AML cells with CBFβ-SMMHC knockdown. RUNX1 binding at one of six sites with RUNX1 occupancy identified by chromatin immunoprecipitation followed by sequencing (RE5, regulatory element 5) was increased in the CBFβ-SMMHC knock-down cells. The RE5 is located 178kb upstream of the NRP1 transcription start site and it is evolutionarily conserved in vertebrates. The deletion of RE5 by gene editing (~50% editing efficiency) increased NRP1 expression 1.8-fold, suggesting that RUNX1 may repress NRP1 expression at by binding to the RE5 enhancer. Taken together, these studies demonstrate that CBFβ-SMMHC regulates expression of cytokine receptors in inv(16) AML. Specifically, it directly regulates expression of the co-receptor NRP1, which is essential for AML survival, acting (at least in part) by regulating FLT3 and TGFB pathways. Disclosures Guzman: SeqRx: Consultancy; BridgeMedicines: Consultancy; Cellectis: Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2021
- Full Text
- View/download PDF
26. A tool compound targeting the core binding factor Runt domain to disrupt binding to CBFβ in leukemic cells
- Author
-
Isaura M. Frost, Roger A. Rajewski, John H. Bushweller, Trista E. North, Charles Schmidt, Anuradha Illendula, Zaw Min Oo, Wanda Kwan, Yunpeng Zhou, Jolanta Grembecka, Virginie Esain, and Nancy A. Speck
- Subjects
0301 basic medicine ,Cancer Research ,Core Binding Factor alpha Subunits ,Core Binding Factor beta Subunit ,Cell growth ,Myeloid leukemia ,Hematology ,medicine.disease ,Core binding factor ,03 medical and health sciences ,Leukemia ,chemistry.chemical_compound ,030104 developmental biology ,Oncology ,RUNX1 ,chemistry ,Cell culture ,hemic and lymphatic diseases ,medicine ,Cancer research - Abstract
The core binding factor (CBF) gene RUNX1 is a target of chromosomal translocations in leukemia, including t(8;21) in acute myeloid leukemia (AML). Normal CBF function is essential for activity of AML1-ETO, product of the t(8;21), and for survival of several leukemias lacking RUNX1 mutations. Using virtual screening and optimization, we developed Runt domain inhibitors which bind to the Runt domain and disrupt its interaction with CBFβ. On-target activity was demonstrated by the Runt domain inhibitors' ability to depress hematopoietic cell formation in zebrafish embryos, reduce growth and induce apoptosis of t(8;21) AML cell lines, and reduce progenitor activity of mouse and human leukemia cells harboring the t(8;21), but not normal bone marrow cells. Runt domain inhibitors had similar effects on murine and human T cell acute lymphocytic leukemia (T-ALL) cell lines. Our results confirmed that Runt domain inhibitors might prove efficacious in various AMLs and in T-ALL.
- Published
- 2017
- Full Text
- View/download PDF
27. Small Molecule Inhibitor of CBFβ-RUNX Binding for RUNX Transcription Factor Driven Cancers
- Author
-
Lauren Mishra, Mahmut Parlak, Adam Boulton, Cem Kuscu, Venkata Sesha Srimath Tirumala, Mazhar Adli, Yunpeng Zhou, John H. Bushweller, Anuradha Illendula, Yan Gao, Anna Pickin, Charles Schmidt, Jane Gilmour, Kevin A. Janes, John Anto Pulikkan, Roger A. Rajewski, Aravinda Kuntimaddi, Jolanta Grembecka, Lucio H. Castilla, Hongliang Zong, Constanze Bonifer, Monica L. Guzman, and Lixin Wang
- Subjects
0301 basic medicine ,Core Binding Factor alpha Subunits ,PPI ,lcsh:Medicine ,Biology ,Core binding factor ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Medicine, General & Internal ,hemic and lymphatic diseases ,medicine ,Triple negative breast cancer ,Transcription factor ,Gene ,Triple-negative breast cancer ,lcsh:R5-920 ,Leukemia ,CBFβ ,lcsh:R ,General Medicine ,medicine.disease ,Molecular biology ,RUNX ,3. Good health ,Cell biology ,030104 developmental biology ,RUNX1 ,chemistry ,Stem cell ,Transcription factor inhibitor ,lcsh:Medicine (General) - Abstract
Transcription factors have traditionally been viewed with skepticism as viable drug targets, but they offer the potential for completely novel mechanisms of action that could more effectively address the stem cell like properties, such as self-renewal and chemo-resistance, that lead to the failure of traditional chemotherapy approaches. Core binding factor is a heterodimeric transcription factor comprised of one of 3 RUNX proteins (RUNX1-3) and a CBFβ binding partner. CBFβ enhances DNA binding of RUNX subunits by relieving auto-inhibition. Both RUNX1 and CBFβ are frequently mutated in human leukemia. More recently, RUNX proteins have been shown to be key players in epithelial cancers, suggesting the targeting of this pathway could have broad utility. In order to test this, we developed small molecules which bind to CBFβ and inhibit its binding to RUNX. Treatment with these inhibitors reduces binding of RUNX1 to target genes, alters the expression of RUNX1 target genes, and impacts cell survival and differentiation. These inhibitors show efficacy against leukemia cells as well as basal-like (triple-negative) breast cancer cells. These inhibitors provide effective tools to probe the utility of targeting RUNX transcription factor function in other cancers.
- Published
- 2016
28. MAPK/ERK2 phosphorylates ERG at serine 283 in leukemic cells and promotes stem cell signatures and cell proliferation
- Author
-
Yizhou Huang, Karen L. MacKenzie, Dominik Beck, Santi Suryani, John E. Pimanda, Vashe Chandrakanthan, Jake Olivier, Adam Boulton, Elias N. Glaros, Richard B. Lock, John H. Bushweller, Julie A. I. Thoms, Shane R. Thomas, Kathy Knezevic, Melinda L. Tursky, and Jason W. H. Wong
- Subjects
0301 basic medicine ,Cancer Research ,genetic structures ,MAP Kinase Signaling System ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Fusion gene ,03 medical and health sciences ,Transcriptional Regulator ERG ,Cell Line, Tumor ,Serine ,Humans ,Protein phosphorylation ,Phosphorylation ,Progenitor cell ,Cell Proliferation ,Binding Sites ,Leukemia ,Myeloid leukemia ,Hematology ,Hematopoietic Stem Cells ,eye diseases ,Leukemia, Myeloid, Acute ,Haematopoiesis ,030104 developmental biology ,Oncology ,Cancer research ,sense organs ,Stem cell ,Transcriptome ,Protein Processing, Post-Translational ,Erg - Abstract
Aberrant ERG (v-ets avian erythroblastosis virus E26 oncogene homolog) expression drives leukemic transformation in mice and high expression is associated with poor patient outcomes in acute myeloid leukemia (AML) and T-acute lymphoblastic leukemia (T-ALL). Protein phosphorylation regulates the activity of many ETS factors but little is known about ERG in leukemic cells. To characterize ERG phosphorylation in leukemic cells, we applied liquid chromatography coupled tandem mass spectrometry and identified five phosphorylated serines on endogenous ERG in T-ALL and AML cells. S283 was distinct as it was abundantly phosphorylated in leukemic cells but not in healthy hematopoietic stem and progenitor cells (HSPCs). Overexpression of a phosphoactive mutant (S283D) increased expansion and clonogenicity of primary HSPCs over and above wild-type ERG. Using a custom antibody, we screened a panel of primary leukemic xenografts and showed that ERG S283 phosphorylation was mediated by mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling and in turn regulated expression of components of this pathway. S283 phosphorylation facilitates ERG enrichment and transactivation at the ERG +85 HSPC enhancer that is active in AML and T-ALL with poor prognosis. Taken together, we have identified a specific post-translational modification in leukemic cells that promotes progenitor proliferation and is a potential target to modulate ERG-driven transcriptional programs in leukemia.
- Published
- 2016
- Full Text
- View/download PDF
29. Abstract 5041: Inhibition of RUNX-CBFβ binding reduces RSPO3 expression and EMT features in breast cancer cells
- Author
-
Lucio H. Castilla, Martín E García Solá, Carla M. Felcher, Edith C. Kordon, John H. Bushweller, and Johanna M. Tocci
- Subjects
Cancer Research ,Gene knockdown ,Mammary tumor ,Wnt signaling pathway ,Cancer ,Biology ,medicine.disease ,Breast cancer ,Oncology ,SOX2 ,Cancer stem cell ,Cancer research ,medicine ,Stem cell - Abstract
We have recently identified R-spondin3 (RSPO3) as a novel key modulator of breast cancer development that may become a potential target for treatment of the basal subtype, which lacks efficient therapeutic options to date. RSPO3 is expressed in the basal stem cell-enriched compartment of mouse mammary glands whereas it is absent in the mature luminal cells. RSPO3 knockdown in mouse mammary tumor cells reduces canonical Wnt signaling pathway, epithelial-to-mesenchymal transition (EMT)-like features, migration capacity, and tumor formation in vivo. To evaluate the relevance of RSPO3 in human breast cancer, we analyzed samples from 75 patients with ductal infiltrating breast carcinomas, where we found a high prevalence of positive immunoreactivity for RSPO3 (70%). Expression data of The Cancer Genome Atlas Network from 1985 human breast carcinoma samples showed that RSPO3 is primarily expressed in Claudin-low breast cancer subtype, which is characterized for having stem cells features. In culture assays revealed that knockdown of RSPO3 expression reduced SOX2 expression and impaired mammary tumor cell ability to form mammospheres. Previous reports suggested the potential involvement of RUNX1-CBFβ on RSPO3 transcription in mammary tumor cells. Therefore, we evaluated if pharmacologic inhibition of RUNX1 activity would reduce RSPO3 expression and activity in the cancer stem cell compartment. Treatment of breast cancer cells with AI-10-104, a small molecule that inhibits CBFβ-RUNX interaction, reduced RSPO3 mRNA and protein levels, and reduced the migration ability of MDA-MB231 breast cancer cells, ability that was recovered upon treatment with recombinant RSPO3. In addition, AI-10-104 inhibited the expression of canonical Wnt pathway targets, EMT and stem-cell markers. Therefore, our results demonstrate that inhibition of RUNX-CBFβ transcriptional activity leads to a down-regulation of RSPO3 expression, constituting a new molecular target for therapeutic modulation in breast cancer treatment. Citation Format: Carla M. Felcher, Johanna M. Tocci, Martin E. Garcia Sola, John H. Bushweller, Lucio H. Castilla, Edith C. Kordon. Inhibition of RUNX-CBFβ binding reduces RSPO3 expression and EMT features in breast cancer cells [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5041.
- Published
- 2020
- Full Text
- View/download PDF
30. RUNX1 mitotically bookmarks target genes that are important for the mammary epithelial-to-mesenchymal transition
- Author
-
Eliana Moskovitz, Andrew J. Fritz, Joseph R. Boyd, Gary S. Stein, Janet L. Stein, John H. Bushweller, Sayyed K. Zaidi, Joshua T. Rose, Jonathan A. R. Gordon, Nicole A. Bouffard, Anuradha Illendula, and Jane B. Lian
- Subjects
0303 health sciences ,Cell ,Biology ,Cell cycle ,Cell morphology ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,hemic and lymphatic diseases ,Gene expression ,embryonic structures ,medicine ,Epithelial–mesenchymal transition ,Gene ,Mitosis ,Transcription factor ,030304 developmental biology - Abstract
RUNX1 has recently been shown to play an important role in determination of mammary epithelial cell identity. However, mechanisms by which loss of the RUNX1 transcription factor in mammary epithelial cells leads to epithelial-to-mesenchymal transition (EMT) are not known. Here, we report mitotic bookmarking of genes by RUNX1 as a potential mechanism to convey regulatory information through successive cell divisions for coordinate control of mammary cell proliferation, growth, and identity. Genome-wide RUNX1 occupancy profiles for asynchronous, mitotically enriched, and early G1 breast epithelial cells reveal RUNX1 is retained during the mitosis to G1 transition on protein coding and long non-coding RNA genes critical for mammary epithelial proliferation, growth, and phenotype maintenance. Disruption of RUNX1 DNA binding and association with mitotic chromosomes alters cell morphology, global protein synthesis, and phenotype-related gene expression. Together, these findings show for the first time that RUNX1 bookmarks a subset of epithelial-related genes during mitosis that remain occupied as cells enter the next cell cycle. Compromising RUNX1 DNA binding initiates EMT, an essential first step in the onset of breast cancer.SignificanceThis study elucidates mitotic gene bookmarking as a potential epigenetic mechanism that impacts breast epithelial cell growth and phenotype and has potential implications in breast cancer onset.
- Published
- 2019
- Full Text
- View/download PDF
31. RUNX proteins desensitize multiple myeloma to lenalidomide via protecting IKZFs from degradation
- Author
-
Luca Bernabei, Alfred L. Garfall, Luca Busino, Laurence Florens, Anita Saraf, Anuradha Illendula, Michael P. Washburn, Xiang Yu Zheng, Nan Zhou, Alvaro Gutierrez-Uzquiza, Dan T. Vogl, and John H. Bushweller
- Subjects
0303 health sciences ,biology ,Chemistry ,Cereblon ,medicine.disease ,IKZF3 ,3. Good health ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.anatomical_structure ,Ubiquitin ,RUNX1 ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,medicine ,Transcription factor ,Sensitization ,Multiple myeloma ,030304 developmental biology ,Lenalidomide ,medicine.drug - Abstract
Ikaros family zinc finger protein 1 and 3 (IKZF1 and IKZF3) are transcription factors that promote multiple myeloma (MM) proliferation. The immunomodulatory imide drug (IMiD) lenalidomide promotes myeloma cell death via Cereblon (CRBN)-dependent ubiquitylation and proteasome-dependent degradation of IKZF1 and IKZF3. Although IMiDs have been used as first-line drugs for MM, the overall survival of refractory MM patients remains poor and demands the identification of novel agents to potentiate the therapeutic effect of IMiDs. Using an unbiased screen based on mass spectrometry, we identified the Runt-related transcription factor 1 and 3 (RUNX1 and RUNX3) as interactors of IKZF1 and IKZF3. Interaction with RUNX1 and RUNX3 inhibits CRBN-dependent binding, ubiquitylation and degradation of IKZF1 and IKZF3 upon lenalidomide treatment. Inhibition of RUNXs, via genetic ablation or a small molecule (AI-10-104), results in sensitization of myeloma cell lines and primary tumors to lenalidomide. Thus, RUNX inhibition represents a valuable therapeutic opportunity to potentiate IMiDs therapy for the treatment of multiple myeloma.
- Published
- 2018
- Full Text
- View/download PDF
32. CBFβ-SMMHC inhibition triggers apoptosis by disrupting MYC chromatin dynamics in acute myeloid leukemia
- Author
-
Jun Yu, John H. Bushweller, Houda Belaghzal, Job Dekker, Jianhong Ou, Carsten Müller-Tidow, Mahesh Hegde, Lihua Julie Zhu, Scot A. Wolfe, Kelsey O’Hagan, Hafiz M. Ahmad, John Anto Pulikkan, Lucio H. Castilla, and Anuradha Illendula
- Subjects
0301 basic medicine ,Myeloid leukemia ,Biology ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Article ,Chromatin ,Cell biology ,BET inhibitor ,03 medical and health sciences ,Leukemia ,chemistry.chemical_compound ,030104 developmental biology ,RUNX1 ,chemistry ,hemic and lymphatic diseases ,medicine ,Enhancer ,Psychological repression ,Transcription factor - Abstract
The fusion oncoprotein CBFβ-SMMHC, expressed in leukemia cases with chromosome 16 inversion, drives leukemia development and maintenance by altering the activity of the transcription factor RUNX1. Here, we demonstrate that CBFβ-SMMHC maintains cell viability by neutralizing RUNX1-mediated repression of MYC expression. Upon pharmacologic inhibition of the CBFβ-SMMHC/RUNX1 interaction, RUNX1 shows increased binding at three MYC distal enhancers, where it represses MYC expression by mediating the replacement of the SWI/SNF complex component BRG1 with the polycomb-repressive complex component RING1B, leading to apoptosis. Combining the CBFβ-SMMHC inhibitor with the BET inhibitor JQ1 eliminates inv(16) leukemia in human cells and a mouse model. Enhancer-interaction analysis indicated that the three enhancers are physically connected with the MYC promoter, and genome-editing analysis demonstrated that they are functionally implicated in deregulation of MYC expression. This study reveals a mechanism whereby CBFβ-SMMHC drives leukemia maintenance and suggests that inhibitors targeting chromatin activity may prove effective in inv(16) leukemia therapy.
- Published
- 2018
33. Tom1 Modulates Binding of Tollip to Phosphatidylinositol 3-Phosphate via a Coupled Folding and Binding Mechanism
- Author
-
Carla V. Finkielstein, Kristen I. Fread, Xiaolin Zhao, Jeffrey F. Ellena, John H. Bushweller, Mary K. Brannon, Daniel G. S. Capelluto, Shuyan Xiao, and Geoffrey S. Armstrong
- Subjects
Models, Molecular ,Proteasome Endopeptidase Complex ,Protein Folding ,Endosome ,Recombinant Fusion Proteins ,Gene Expression ,Plasma protein binding ,Endosomes ,Biology ,Crystallography, X-Ray ,Protein Structure, Secondary ,chemistry.chemical_compound ,Protein structure ,Phosphatidylinositol Phosphates ,Structural Biology ,Escherichia coli ,Humans ,Phosphatidylinositol ,Binding site ,Molecular Biology ,C2 domain ,Binding Sites ,Ubiquitin ,TOLLIP ,Phosphatidylinositol 3-phosphate ,Intracellular Signaling Peptides and Proteins ,Ubiquitination ,Proteins ,Cell biology ,Protein Structure, Tertiary ,Protein Transport ,chemistry ,HeLa Cells ,Protein Binding - Abstract
SummaryEarly endosomes represent the first sorting station for vesicular ubiquitylated cargo. Tollip, through its C2 domain, associates with endosomal phosphatidylinositol 3-phosphate (PtdIns(3)P) and binds ubiquitylated cargo in these compartments via its C2 and CUE domains. Tom1, through its GAT domain, is recruited to endosomes by binding to the Tollip Tom1-binding domain (TBD) through an unknown mechanism. Nuclear magnetic resonance data revealed that Tollip TBD is a natively unfolded domain that partially folds at its N terminus when bound to Tom1 GAT through high-affinity hydrophobic contacts. Furthermore, this association abrogates binding of Tollip to PtdIns(3)P by additionally targeting its C2 domain. Tom1 GAT is also able to bind ubiquitin and PtdIns(3)P at overlapping sites, albeit with modest affinity. We propose that association with Tom1 favors the release of Tollip from endosomal membranes, allowing Tollip to commit to cargo trafficking.
- Published
- 2015
- Full Text
- View/download PDF
34. Degree of Recruitment of DOT1L to MLL-AF9 Defines Level of H3K79 Di- and Tri-methylation on Target Genes and Transformation Potential
- Author
-
Nicholas J. Achille, Aravinda Kuntimaddi, Nancy J. Zeleznik-Le, Jeremy Thorpe, Alyson A. Lokken, John H. Bushweller, Charles S. Hemenway, Ritambhara Singh, and Mazhar Adli
- Subjects
Chromatin Immunoprecipitation ,Magnetic Resonance Spectroscopy ,Methyltransferase ,Oncogene Proteins, Fusion ,Molecular Sequence Data ,Chromosomal translocation ,Biology ,Methylation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Histones ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Humans ,Point Mutation ,Amino Acid Sequence ,Gene ,neoplasms ,lcsh:QH301-705.5 ,030304 developmental biology ,Genetics ,0303 health sciences ,Binding Sites ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,Sequence Analysis, DNA ,DOT1L ,Recombinant Proteins ,Protein Structure, Tertiary ,Histone ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,biology.protein ,Myeloid-Lymphoid Leukemia Protein ,Chromatin immunoprecipitation ,Protein Binding - Abstract
SummaryThe MLL gene is a common target of chromosomal translocations found in human leukemia. MLL-fusion leukemia has a consistently poor outcome. One of the most common translocation partners is AF9 (MLLT3). MLL-AF9 recruits DOT1L, a histone 3 lysine 79 methyltransferase (H3K79me1/me2/me3), leading to aberrant gene transcription. We show that DOT1L has three AF9 binding sites and present the nuclear magnetic resonance (NMR) solution structure of a DOT1L-AF9 complex. We generate structure-guided point mutations and find that they have graded effects on recruitment of DOT1L to MLL-AF9. Chromatin immunoprecipitation sequencing (ChIP-seq) analyses of H3K79me2 and H3K79me3 show that graded reduction of the DOT1L interaction with MLL-AF9 results in differential loss of H3K79me2 and me3 at MLL-AF9 target genes. Furthermore, the degree of DOT1L recruitment is linked to the level of MLL-AF9 hematopoietic transformation.
- Published
- 2015
35. A small-molecule inhibitor of the aberrant transcription factor CBFβ-SMMHC delays leukemia in mice
- Author
-
Roger A. Rajewski, Hongliang Zong, Adam Boulton, Yunpeng Zhou, John Anto Pulikkan, Aravinda Kuntimaddi, Anuradha Illendula, John H. Bushweller, Yan Gao, Liting Xue, Monica L. Guzman, Siddhartha Sen, Jolanta Grembecka, and Lucio H. Castilla
- Subjects
Myeloid ,Oncogene Proteins, Fusion ,Antineoplastic Agents ,Biology ,Core binding factor ,Article ,Small Molecule Libraries ,Mice ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Interaction Maps ,Transcription factor ,Multidisciplinary ,Myeloid leukemia ,medicine.disease ,Fusion protein ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,RUNX1 ,chemistry ,Core Binding Factor Alpha 2 Subunit ,Immunology ,Cancer research ,Benzimidazoles ,Female - Abstract
Toward drugging the undruggable in cancer Many human cancers are characterized by inappropriate activity of transcription factors. These proteins are attractive drug targets in principle, but normalizing their function requires drugs that modulate specific protein-protein interactions, a goal that has been challenging. In acute myeloid leukemia, a chromosomal translocation creates an aberrant form of the transcription factor CBF-beta, which outcompetes “normal” CBF-beta for binding to another transcription factor called RUNX1, thereby deregulating its activity. Illendula et al. identified and chemically optimized a small molecule that selectively disrupts the interaction between the aberrant CBF-beta and RUNX1 (see the Perspective by Koehler and Chen). This molecule restored normal gene expression patterns and delayed leukemia progression in mice. Thus, transcription factors may not be as undruggable as once thought. Science , this issue p. 779 ; see also p. 713
- Published
- 2015
- Full Text
- View/download PDF
36. RUNX1 is required for oncogenic
- Author
-
Anuradha Illendula, John H. Bushweller, Jessica Tesell, John Anto Pulikkan, Justine E. Roderick, Jun Yu, Nicole Hermance, Lucio H. Castilla, Lihua Julie Zhu, Michelle A. Kelliher, and AHyun Choi
- Subjects
0301 basic medicine ,LMO2 ,Carcinogenesis ,Apoptosis ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Basic Helix-Loop-Helix Transcription Factors ,MYB ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Cell Line, Transformed ,Lymphoid Neoplasia ,Receptors, Notch ,Gene Expression Regulation, Leukemic ,Hematology ,Chromatin ,Leukemia ,Enhancer Elements, Genetic ,RUNX1 ,030220 oncology & carcinogenesis ,embryonic structures ,Core Binding Factor Alpha 2 Subunit ,Protein Binding ,Cell Survival ,Immunology ,Biology ,Core Binding Factor beta Subunit ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Proto-Oncogene Proteins c-myb ,Cell Line, Tumor ,Proto-Oncogene Proteins ,medicine ,Gene silencing ,Animals ,Humans ,Transcription factor ,Cell Proliferation ,Cell Biology ,Oncogenes ,medicine.disease ,030104 developmental biology ,chemistry ,Cancer research ,Gene Deletion ,TAL1 - Abstract
The gene encoding the RUNX1 transcription factor is mutated in a subset of T-cell acute lymphoblastic leukemia (T-ALL) patients, and RUNX1 mutations are associated with a poor prognosis. These mutations cluster in the DNA-binding Runt domain and are thought to represent loss-of-function mutations, indicating that RUNX1 suppresses T-cell transformation. RUNX1 has been proposed to have tumor suppressor roles in T-cell leukemia homeobox 1/3-transformed human T-ALL cell lines and NOTCH1 T-ALL mouse models. Yet, retroviral insertional mutagenesis screens identify RUNX genes as collaborating oncogenes in MYC-driven leukemia mouse models. To elucidate RUNX1 function(s) in leukemogenesis, we generated Tal1/Lmo2/Rosa26-CreERT2Runx1f/f mice and examined leukemia progression in the presence of vehicle or tamoxifen. We found that Runx1 deletion inhibits mouse leukemic growth in vivo and that RUNX silencing in human T-ALL cells triggers apoptosis. We demonstrate that a small molecule inhibitor, designed to interfere with CBFβ binding to RUNX proteins, impairs the growth of human T-ALL cell lines and primary patient samples. We demonstrate that a RUNX1 deficiency alters the expression of a crucial subset of TAL1- and NOTCH1-regulated genes, including the MYB and MYC oncogenes, respectively. These studies provide genetic and pharmacologic evidence that RUNX1 has oncogenic roles and reveal RUNX1 as a novel therapeutic target in T-ALL.
- Published
- 2017
37. Molecular Basis and Targeted Inhibition of CBFβ-SMMHC Acute Myeloid Leukemia
- Author
-
Lucio H, Castilla and John H, Bushweller
- Subjects
Leukemia, Myeloid, Acute ,Myosin Heavy Chains ,Oncogene Proteins, Fusion ,Animals ,Humans ,Core Binding Factor beta Subunit - Abstract
Acute myeloid leukemia (AML) is characterized by recurrent chromosomal rearrangements that encode for fusion proteins which drive leukemia initiation and maintenance. The inv(16) (p13q22) rearrangement is a founding mutation and the associated CBFβ-SMMHC fusion protein is essential for the survival of inv(16) AML cells. This Chapter will discuss our understanding of the function of this fusion protein in disrupting hematopoietic homeostasis and creating pre-leukemic blasts, in its cooperation with other co-occurring mutations during leukemia initiation, and in leukemia maintenance. In addition, this chapter will discuss the current approaches used for the treatment of inv(16) AML and the recent development of AI-10-49, a selective targeted inhibitor of CBFβ-SMMHC/RUNX1 binding, the first candidate targeted therapy for inv(16) AML.
- Published
- 2017
38. Importance of a specific amino acid pairing for murine MLL leukemias driven by MLLT1/3 or AFF1/4
- Author
-
Bhavna Malik, Shubin Zhang, Nicholas J. Achille, John H. Bushweller, Alyson A. Lokken, Charles S. Hemenway, Jeffrey J. Lin, Benjamin I. Leach, Aravinda Kuntimaddi, Ming-Jin Chang, Nancy J. Zeleznik-Le, and Jacqueline Nesbit
- Subjects
Cancer Research ,Molecular Sequence Data ,Plasma protein binding ,Biology ,DNA-binding protein ,Article ,Mice ,hemic and lymphatic diseases ,Animals ,Humans ,Amino Acid Sequence ,Amino Acids ,Peptide sequence ,Transcription factor ,chemistry.chemical_classification ,Messenger RNA ,Leukemia, Experimental ,Sequence Homology, Amino Acid ,HEK 293 cells ,Nuclear Proteins ,Hematology ,Fusion protein ,Molecular biology ,Amino acid ,Cell biology ,DNA-Binding Proteins ,HEK293 Cells ,Oncology ,chemistry ,Transcriptional Elongation Factors ,Transcription Factors - Abstract
Acute leukemias caused by translocations of the MLL gene at chromosome 11 band q23 (11q23) are characterized by a unique gene expression profile. More recently, data from several laboratories indicate that the most commonly encountered MLL fusion proteins, MLLT1, MLLT3, and AFF1 are found within a molecular complex that facilitates the elongation phase of mRNA transcription. Mutational analyses suggest that interaction between the MLLT1/3 proteins and AFF family proteins are required for experimental transformation of hematopoietic progenitor cells (HPCs). Here, we define a specific pairing of two amino acids that creates a salt bridge between MLLT1/3 and AFF proteins that is critically important for MLL-mediated transformation of HPCs. Our findings, coupled with the newly defined structure of MLLT3 in complex with AFF1, should facilitate the development of small molecules that block this amino acid interaction and interfere with the activity of the most common MLL oncoproteins.
- Published
- 2014
- Full Text
- View/download PDF
39. EXPLOITING CBF DEPENDENCY IN B-ACUTE LYMPHOBLASTIC LEUKAEMIA AS A NOVEL THERAPEUTIC APPROACH
- Author
-
Elitza Deltcheva, Anuradha Illendula, Jyoti Bikram Chettri, Chela James, Simon Richardson, John H. Bushweller, Rachael Nimmo, Charlotta Böiers, Jason Wray, and Tariq Enver
- Subjects
Cancer Research ,Chromosomal translocation ,Cell Biology ,Hematology ,Cell cycle ,Biology ,ETV6 ,chemistry.chemical_compound ,medicine.anatomical_structure ,Downregulation and upregulation ,RUNX1 ,chemistry ,hemic and lymphatic diseases ,embryonic structures ,Genetics ,medicine ,Cancer research ,Mass cytometry ,E2F ,Molecular Biology ,B cell - Abstract
Leukaemia is the most common childhood cancer and acute lymphoblastic leukaemia (ALL) is responsible for a third of all childhood cancer deaths. Despite the overall good prognosis, failure to eradicate the disease leads to relapse in ∼20% of patients and survival rates for infants are less than 50%. There is therefore a clear unmet need for less toxic and more effective targeted treatments. ETV6-RUNX1 translocation is the first-hit event in ∼25% of B-ALLs, initiating a clinically silent pre-leukaemia in utero. The translocation fuses almost the entire RUNX1 protein to the repressor domain of ETV6 and the chimeric product is proposed to mediate a block in B cell differentiation by repressing RUNX1 targets. Interestingly, the remaining RUNX1 allele is not mutated, but is instead often amplified suggesting an onco-supportive role of the endogenous RUNX1. Using genetic and pharmacological inhibition, we have discovered that RUNX1/CBF is essential for the growth and survival of B-ALL cells in vitro and in vivo. Global RNA-Seq on CBF-depleted B-ALL cells revealed significant downregulation of E2F and Myc signature genes along with a marked increase of p53 targets, consistent with these cells accumulating in G0 and subsequently becoming apoptotic. Notably, RNA-Seq and mass cytometry analyses of pro-B cells derived from an hiPSC ETV6-RUNX1 knockin model reveal a similar cell cycle trend indicating that ETV6-RUNX1-mediated inhibition of RUNX1 targets may induce a quiescence-like pre-leukaemic state while continued RUNX1 expression is essential for the survival of overt leukaemia. We propose that CBF-dependency is an Achilles heel of ALL cells that could be exploited by targeting either CBF itself or its key downstream nodes. By combining clinically tested compounds targeting multiple CBF effectors we aim to improve efficacy of potential treatments and circumvent resistance.
- Published
- 2019
- Full Text
- View/download PDF
40. Molecular Basis and Targeted Inhibition of CBFβ-SMMHC Acute Myeloid Leukemia
- Author
-
Lucio H. Castilla and John H. Bushweller
- Subjects
0301 basic medicine ,Mutation ,medicine.medical_treatment ,Myeloid leukemia ,biochemical phenomena, metabolism, and nutrition ,Biology ,medicine.disease_cause ,medicine.disease ,Fusion protein ,Targeted therapy ,03 medical and health sciences ,AI-10-49 ,chemistry.chemical_compound ,Leukemia ,Haematopoiesis ,030104 developmental biology ,chemistry ,RUNX1 ,hemic and lymphatic diseases ,medicine ,Cancer research - Abstract
Acute myeloid leukemia (AML) is characterized by recurrent chromosomal rearrangements that encode for fusion proteins which drive leukemia initiation and maintenance. The inv(16) (p13q22) rearrangement is a founding mutation and the associated CBFβ-SMMHC fusion protein is essential for the survival of inv(16) AML cells. This Chapter will discuss our understanding of the function of this fusion protein in disrupting hematopoietic homeostasis and creating pre-leukemic blasts, in its cooperation with other co-occurring mutations during leukemia initiation, and in leukemia maintenance. In addition, this chapter will discuss the current approaches used for the treatment of inv(16) AML and the recent development of AI-10-49, a selective targeted inhibitor of CBFβ-SMMHC/RUNX1 binding, the first candidate targeted therapy for inv(16) AML.
- Published
- 2017
- Full Text
- View/download PDF
41. Structure and Biophysics of CBFβ/RUNX and Its Translocation Products
- Author
-
Tahir H. Tahirov and John H. Bushweller
- Subjects
0301 basic medicine ,030102 biochemistry & molecular biology ,Chemistry ,Protein subunit ,Chromosomal translocation ,Core binding factor ,Fusion protein ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,RUNX1 ,A-DNA ,Protein activity ,Transcription factor - Abstract
The core binding factor (CBF) transcription factor is somewhat unique in that it is composed of a DNA binding RUNX subunit (RUNX1, 2, or 3) and a non-DNA binding CBFβ subunit, which modulates RUNX protein activity by modulating the auto-inhibition of the RUNX subunits. Since the discovery of this fascinating transcription factor more than 20 years ago, there has been a robust effort to characterize the structure as well as the biochemical properties of CBF. More recently, these efforts have also extended to the fusion proteins that arise from the subunits of CBF in leukemia. This chapter highlights the work of numerous labs which has provided a detailed understanding of the structure and function of this transcription factor and its fusion proteins.
- Published
- 2017
- Full Text
- View/download PDF
42. Direct Binding of BCOR, but Not CBX8, to MLL-AF9 Is Essential for MLL-AF9 Leukemia Via Regulation of the EYA1/SIX1 Gene Network
- Author
-
Nancy J. Zeleznik-Le, Nicholas J. Achille, Charles Schmidt, Adam Boulton, Shubin Zhang, John H. Bushweller, Benjamin I. Leach, and Aravinda Kuntimaddi
- Subjects
0301 basic medicine ,Histone H3 Lysine 4 ,Immunology ,Context (language use) ,Cell Biology ,Hematology ,DOT1L ,Biology ,HDAC3 ,Biochemistry ,Cell biology ,03 medical and health sciences ,Histone H3 ,030104 developmental biology ,0302 clinical medicine ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Histone methyltransferase ,Transcriptional regulation ,H3K4me3 - Abstract
The mixed lineage leukemia (MLL) protein is a histone methyltransferase that writes the histone H3 lysine 4 trimethyl (H3K4me3) mark at the promoters of target genes such as HOXA9 and MEIS1. MLL is the target of chromosomal translocations that fuse it in frame to one of over 90 partners, leading to acute myeloid and lymphoid leukemias (AML and ALL, respectively) characterized by poor prognoses1. MLL fusions activate transcription by recruiting the AF4 family/ENL family/P-TEFb (AEP) complex and the DOT1L-AF10 family-ENL family complex (DOT1L complex or DotCom). Transcriptional activation via AF4 recruitment and transcriptional maintenance via DOT1L recruitment are required for MLL leukemias. Despite the large number of fusion partners, members of the AEP complex account for nearly 70% of MLL rearrangements1. These fusions constitutively activate MLL targets by bypassing recruitment via ENL (MLLT1) and AF9 (MLLT3) YEATS domain binding to crotonylated or acetylated histone H3. The AF9 ANC1 homology domain (AHD), retained in MLL fusions, is intrinsically disordered, but undergoes coupled folding and binding upon interaction with its binding proteins2. The AHD recruits AF4 and DOT1L, which support transcriptional elongation, as well as the BCL6 corepressor (BCOR) and chromobox homolog 8 (CBX8), which are implicated in transcriptional repression. CBX8 (HPC3) is a mammalian ortholog of Drosophila polycomb that binds trimethylated histone H3 lysine 9 and 27 (H3K9me3 and H3K27me3) with variable affinity. Previous reports indicate CBX8 is required for MLL-AF9 and MLL-ENL. BCOR is a transcriptional corepressor that augments BCL6-mediated repression. The BCL6 POZ domain forms a ternary complex with BCOR and SMRT, repressing targets via recruitment of PRC1.1 and HDAC3. BCOR translocations and mutations have been found in a range of cancers. Although it is broadly expressed throughout the hematopoietic system (Bloodspot), little is known about BCOR function in hematopoiesis. Recently, BCOR was shown to have a role in maintenance of human embryonic stem cell pluripotency. BCOR has also been implicated in regulation of myeloid cell proliferation and differentiation and is necessary for MLL-AF9 leukemogenesis. While the roles of the direct MLL-AF9/AF4 and MLL-AF9/DOT1L interactions have been the subject of previous structural and functional studies2-4, the roles of the direct interactions of MLL-AF9 with CBX8 and BCOR remain relatively uncharacterized. We determined the structures of the AF9 AHD-CBX8 and AF9 AHD-BCOR complexes. Based on the structures, we developed point mutants to increase and decrease affinity of CBX8 for AF9. Increased affinity decreased colony forming ability and induced differentiation of MLL-AF9-transformed cells, while decreased affinity had no effect. An additional point mutant was developed to selectively disrupt BCOR binding to AF9. In the context of MLL-AF9, this mutant increases proliferative ability without an effect on colony formation and is unable to cause leukemia in vivo. RNAseq analysis reveals that this mutant affects a different set of genes than loss of DOT1L or AF4 binding or gain of CBX8 binding, leading to a phenotype distinct from that seen with perturbation of other AF9 interactions, functionally distinguishing proliferative capacity from in vivo leukemogenesis. In particular, substantial effects were observed on EYA1 expression, suggesting a critical role for the EYA1/SIX gene network in MLL-AF9 leukemia. 1 Meyer, C. et al. The MLL recombinome of acute leukemias in 2017. Leukemia32, 273-284, doi:10.1038/leu.2017.213 (2018). 2 Leach, B. I. et al. Leukemia fusion target AF9 is an intrinsically disordered transcriptional regulator that recruits multiple partners via coupled folding and binding. Structure21, 176-183, doi:10.1016/j.str.2012.11.011 (2013). 3 Kuntimaddi, A. et al. Degree of recruitment of DOT1L to MLL-AF9 defines level of H3K79 Di- and tri-methylation on target genes and transformation potential. Cell reports11, 808-820, doi:10.1016/j.celrep.2015.04.004 (2015). 4 Lokken, A. A. et al. Importance of a specific amino acid pairing for murine MLL leukemias driven by MLLT1/3 or AFF1/4. Leukemia research38, 1309-1315, doi:10.1016/j.leukres.2014.08.010 (2014). Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
43. The PHD3 Domain of MLL Acts as a CYP33-Regulated Switch between MLL-Mediated Activation and Repression
- Author
-
Gayathree Raman, Rebecca H. Schwantes, Sangho Park, John H. Bushweller, Manuel O. Diaz, and Ute Osmers
- Subjects
Down-Regulation ,Repressor ,Biology ,Biochemistry ,Article ,Histones ,Cyclophilins ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,hemic and lymphatic diseases ,Humans ,Histone H3 acetylation ,Psychological repression ,030304 developmental biology ,Regulation of gene expression ,Peptidylprolyl isomerase ,0303 health sciences ,Binding Sites ,Peptidylprolyl Isomerase ,Molecular biology ,Protein Structure, Tertiary ,Up-Regulation ,Repressor Proteins ,Histone ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,biology.protein ,H3K4me3 ,Myeloid-Lymphoid Leukemia Protein ,Protein Binding - Abstract
The mixed lineage leukemia (MLL) gene plays a critical role in epigenetic regulation of gene expression and is a frequent target of chromosomal translocations leading to leukemia. MLL plant homeodomain 3 (PHD3) is lost in all MLL translocation products, and reinsertion of PHD3 into MLL fusion proteins abrogates their transforming activity. PHD3 has been shown to interact with the RNA-recognition motif (RRM) domain of human nuclear Cyclophilin33 (CYP33). Here, we show that CYP33 mediates downregulation of the expression of MLL target genes HOXC8, HOXA9, CDKN1B, and C-MYC, in a proline isomerase-dependent manner. This downregulation correlates with the reduction of trimethylated lysine 4 of histone H3 (H3K4me3) and histone H3 acetylation. We have structurally characterized both the PHD3 and CYP33 RRM domains and analyzed their binding to one another. The PHD3 domain binds H3K4me3 (preferentially) and the CYP33 RRM domain at distinct sites. Our binding data show that binding of H3K4me3 to PHD3 and binding of the CYP33 RRM domain to PHD3 are mutually inhibitory, implying that PHD3 is a molecular switch for the transition between activation and repression of target genes. To explore the possible mechanism of CYP33/PHD3-mediated repression, we have analyzed the CYP33 proline isomerase activity on various H3 and H4 peptides and shown selectivity for two sites in H3. Our results provide a possible mechanism for the MLL PHD3 domain to act as a switch between activation and repression.
- Published
- 2010
- Full Text
- View/download PDF
44. Accelerated Leukemogenesis by Truncated CBFβ-SMMHC Defective in High-Affinity Binding with RUNX1
- Author
-
John H. Bushweller, Gang Huang, Sheila T. Compton, Ling Zhao, Yoshiaki Ito, Thomas A. Paul, Takeshi Corpora, R. Katherine Hyde, Linda Wolff, Mondira Kundu, Lisa Garrett, James C. Mulloy, Pengfei Liu, Mark Wunderlich, and Yasuhiko Kamikubo
- Subjects
Cancer Research ,Oncogene Proteins, Fusion ,Mice, Transgenic ,CELLCYCLE ,Plasma protein binding ,Biology ,Article ,Core Binding Factor beta Subunit ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Phosphorylation ,030304 developmental biology ,0303 health sciences ,Leukemia, Experimental ,Myeloid leukemia ,Cell Biology ,medicine.disease ,Molecular biology ,Fusion protein ,Cell biology ,Leukemia ,Haematopoiesis ,Oncology ,RUNX1 ,chemistry ,030220 oncology & carcinogenesis ,Core Binding Factor Alpha 2 Subunit ,embryonic structures ,Protein Binding ,Binding domain - Abstract
Dominant RUNX1 inhibition has been proposed as a common pathway for CBF leukemia. CBF beta-SMMHC, a fusion protein in human acute myeloid leukemia (AML), dominantly inhibits RUNX1 largely through its RUNX1 high-affinity binding domain (HABD). However, the type I CBF beta-SMMHC fusion in AML patients lacks HABD. Here, we report that the type I CBF beta-SMMHC protein binds RUNX1 inefficiently. Knockin mice expressing CBF beta-SMMHC with a HABD deletion developed leukemia quickly, even though hematopoietic defects associated with Runx1-inhibition were partially rescued. A larger pool of leukemia-initiating cells, increased MN1 expression, and retention of RUNX1 phosphorylation are potential mechanisms for accelerated leukemia development in these mice. Our data suggest that RUNX1 dominant inhibition may not be a critical step for leukemogenesis by CBF beta-SMMHC.
- Published
- 2010
- Full Text
- View/download PDF
45. The solution structure and dynamics of the DH-PH module of PDZRhoGEF in isolation and in complex with nucleotide-free RhoA
- Author
-
Urszula Derewenda, Jakub Gruszczyk, Aming Zhang, Zygmunt S. Derewenda, Dmitri I. Svergun, Tomasz Cierpicki, John H. Bushweller, Marta Kasterka, Meiying Zheng, Jakub Bielnicki, Maxim V. Petoukhov, and Erik J. Fernandez
- Subjects
chemistry.chemical_classification ,RHOA ,GTP' ,biology ,Chemistry ,Protein dynamics ,GTPase ,Crystal structure ,Biochemistry ,Crystallography ,Protein structure ,biology.protein ,Nucleotide ,Guanine nucleotide exchange factor ,Molecular Biology - Abstract
The DH-PH domain tandems of Dbl-homology guanine nucleotide exchange factors catalyze the exchange of GTP for GDP in Rho-family GTPases, and thus initiate a wide variety of cellular signaling cascades. Although several crystal structures of complexes of DH-PH tandems with cognate, nucleotide free Rho GTPases are known, they provide limited information about the dynamics of the complex and it is not clear how accurately they represent the structures in solution. We used a complementary combination of nuclear magnetic resonance (NMR), small-angle X-ray scattering (SAXS), and hydrogen-deuterium exchange mass spectrometry (DXMS) to study the solution structure and dynamics of the DH-PH tandem of RhoA-specific exchange factor PDZRhoGEF, both in isolation and in complex with nucleotide free RhoA. We show that in solution the DH-PH tandem behaves as a rigid entity and that the mutual disposition of the DH and PH domains remains identical within experimental error to that seen in the crystal structure of the complex, thus validating the latter as an accurate model of the complex in vivo. We also show that the nucleotide-free RhoA exhibits elevated dynamics when in complex with DH-PH, a phenomenon not observed in the crystal structure, presumably due to the restraining effects of crystal contacts. The complex is readily and rapidly dissociated in the presence of both GDP and GTP nucleotides, with no evidence of intermediate ternary complexes.
- Published
- 2009
- Full Text
- View/download PDF
46. Structure of the AML1-ETO eTAFH domain–HEB peptide complex and its contribution to AML1-ETO activity
- Author
-
Marco Tonelli, Sangho Park, Xiongwei Cai, John H. Bushweller, Tomasz Cierpicki, Nancy A. Speck, and Wei Chen
- Subjects
Male ,Oncogene Proteins, Fusion ,Cellular differentiation ,Immunology ,Peptide ,Biology ,Biochemistry ,DNA-binding protein ,Protein Structure, Secondary ,Proto-Oncogene Proteins c-myc ,Mice ,Structure-Activity Relationship ,Protein structure ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Chlorocebus aethiops ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Nuclear Magnetic Resonance, Biomolecular ,neoplasms ,Transcription factor ,Cells, Cultured ,chemistry.chemical_classification ,Myeloid Neoplasia ,COS cells ,Cell Differentiation ,Cell Biology ,Hematology ,Molecular biology ,Fusion protein ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,chemistry ,COS Cells ,Core Binding Factor Alpha 2 Subunit ,STAT6 Transcription Factor ,Cell Division ,Granulocytes ,Transcription Factors - Abstract
AML1-ETO is the chimeric protein product of the t(8;21) in acute myeloid leukemia. The ETO portion of the fusion protein includes the eTAFH domain, which is homologous to several TATA binding protein–associated factors (TAFs) and interacts with E proteins (E2A and HEB). It has been proposed that AML1-ETO–mediated silencing of E protein function might be important for t(8;21) leukemogenesis. Here, we determined the solution structure of a complex between the AML1-ETO eTAFH domain and an interacting peptide from HEB. On the basis of the structure, key residues in AML1-ETO for HEB association were mutated. These mutations do not impair the ability of AML1-ETO to enhance the clonogenic capacity of primary mouse bone marrow cells and do not eliminate its ability to repress proliferation or granulocyte differentiation. Therefore, the eTAFH-E protein interaction appears to contribute relatively little to the activity of AML1-ETO.
- Published
- 2009
- Full Text
- View/download PDF
47. MLL protects CpG clusters from methylation within the Hoxa9 gene, maintaining transcript expression
- Author
-
Catherine Theisler, John H. Bushweller, Frank Erfurth, Relja Popovic, Tomasz Cierpicki, Tara Stuart, Manuel O. Diaz, Zhen Biao Xia, Nancy J. Zeleznik-Le, and Jolanta Grembecka
- Subjects
Homeodomain Proteins ,Genetics ,Regulation of gene expression ,Multidisciplinary ,Methyltransferase ,Transcription, Genetic ,Methylation ,DNA Methylation ,Biological Sciences ,Biology ,Cell Line ,Mice ,Epigenetics of physical exercise ,Gene Expression Regulation ,CpG site ,hemic and lymphatic diseases ,DNA methylation ,Animals ,Humans ,CpG Islands ,Epigenetics ,neoplasms ,Gene ,Myeloid-Lymphoid Leukemia Protein - Abstract
Homeobox ( HOX ) genes play a definitive role in determination of cell fate during embryogenesis and hematopoiesis. MLL -related leukemia is coincident with increased expression of a subset of HOX genes, including HOXA9 . MLL functions to maintain, rather than initiate, expression of its target genes. However, the mechanism of MLL maintenance of target gene expression is not understood. Here, we demonstrate that Mll binds to specific clusters of CpG residues within the Hoxa9 locus and regulates expression of multiple transcripts. The presence of Mll at these clusters provides protection from DNA methylation. shRNA knock-down of Mll reverses the methylation protection status at the previously protected CpG clusters; methylation at these CpG residues is similar to that observed in Mll null cells. Furthermore, reconstituting MLL expression in Mll null cells can reverse DNA methylation of the same CpG residues, demonstrating a dominant effect of MLL in protecting this specific region from DNA methylation. Intriguingly, an oncogenic MLL-AF4 fusion can also reverse DNA methylation, but only for a subset of these CpGs. This method of transcriptional regulation suggests a mechanism that explains the role of Mll in transcriptional maintenance, but it may extend to other CpG DNA binding proteins. Protection from methylation may be an important mechanism of epigenetic inheritance by regulating the function of both de novo and maintenance DNA methyltransferases.
- Published
- 2008
- Full Text
- View/download PDF
48. Structural and Functional Basis of Alternative ESCRT-0 Protein Complexes
- Author
-
Xiaolin Zhao, Carla V. Finkielstein, Jeffrey Ellena, Mary K. Brannon, Kristen Fread, Wen Xiong, Daniel G. S. Capelluto, John H. Bushweller, Shuyan Xiao, and Geoffrey Armstrong
- Subjects
biology ,Endosome ,TOLLIP ,Biophysics ,ESCRT ,Cell biology ,chemistry.chemical_compound ,Ubiquitin ,chemistry ,Biochemistry ,biology.protein ,Phosphatidylinositol ,Function (biology) ,Intracellular ,C2 domain - Abstract
Early endosomes represent the first sorting station for vesicular ubiquitylated cargo. Cargo transport is mediated by the endosomal sorting complex required for transport (ESCRT) machinery. Similar to the structural organization of ESCRT-0 proteins, alternative ESCRT-0 (alt-ESCRT-0) proteins, such as Tollip and Tom1, also present multiple ubiquitin-binding domains, including the C2 and CUE (Tollip) and VHS and GAT (Tom1) domains. Tollip localizes the Tollip-Tom1 complex at endosomal compartments by association with phosphatidylinositol 3-phosphate (PtdIns(3)P) through its central C2 domain. Tom1, through its GAT domain, is recruited to endosomes by binding to Tollip's Tom1-binding domain (TBD) through an unknown mechanism. Our NMR data revealed that Tollip TBD is a natively unfolded domain that partially folds at its N-terminus when bound to the first two helices of the Tom1 GAT domain through high affinity hydrophobic contacts. Furthermore, this association abrogates binding of Tollip to PtdIns(3)P by additionally targeting its C2 domain. Binding of the Tollip C2 domain is mediated by the third helix of the Tom1 GAT domain. We also show that the Tom1 GAT domain is able to bind ubiquitin and PtdIns(3)P at overlapping sites, albeit with modest affinity. We propose that association with Tom1 favors Tollip's release from endosomal membranes, allowing Tollip to commit to cargo trafficking. More recently, Tom1 has been shown to associate to signaling endosomal PtdIns(5)P through its VHS domain delaying epidermal growth factor receptor degradation in a bacterial infection model. We found that PtdIns(5)P binding of the Tom1 VHS domain is in the moderately fast-exchange regime at the NMR timescale. We propose that an incremental change in PtdIns(5)P intracellular levels sequesters Tom1 in endosomal cargo-free membrane domains, keeping the protein from its cargo sorting function.
- Published
- 2016
- Full Text
- View/download PDF
49. Allosteric Inhibition of the Protein-Protein Interaction between the Leukemia-Associated Proteins Runx1 and CBFβ
- Author
-
John H. Bushweller, Michael J. Gorczynski, Takeshi Corpora, Mohini Sridharan, Liya Roudaia, Michael G. Douvas, Ryan H. Lilien, Jolanta Grembecka, Nancy A. Speck, Miki Newman, Izabela Bielnicka, Milton L. Brown, Bruce R. Donald, Yunpeng Zhou, Yali Kong, Jianxia Shi, and Gwen B. Baber
- Subjects
Magnetic Resonance Spectroscopy ,Recombinant Fusion Proteins ,Allosteric regulation ,Clinical Biochemistry ,Biology ,Core binding factor ,Biochemistry ,Core Binding Factor beta Subunit ,Translocation, Genetic ,Protein–protein interaction ,chemistry.chemical_compound ,Cell Line, Tumor ,hemic and lymphatic diseases ,Myosin ,Drug Discovery ,Fluorescence Resonance Energy Transfer ,Humans ,Enzyme Inhibitors ,Molecular Biology ,Cell Proliferation ,Pharmacology ,Binding Sites ,Leukemia ,Myosin Heavy Chains ,Smooth Muscle Myosins ,General Medicine ,Fusion protein ,Hematopoiesis ,A-site ,CHEMBIO ,RUNX1 ,chemistry ,SIGNALING ,Docking (molecular) ,Core Binding Factor Alpha 2 Subunit ,embryonic structures ,Molecular Medicine ,Allosteric Site - Abstract
The two subunits of core binding factor (Runx1 and CBFbeta) play critical roles in hematopoiesis and are frequent targets of chromosomal translocations found in leukemia. The binding of the CBFbeta-smooth muscle myosin heavy chain (SMMHC) fusion protein to Runx1 is essential for leukemogenesis, making this a viable target for treatment. We have developed inhibitors with low micromolar affinity which effectively block binding of Runx1 to CBFbeta. NMR-based docking shows that these compounds bind to CBFbeta at a site displaced from the binding interface for Runx1, that is, these compounds function as allosteric inhibitors of this protein-protein interaction, a potentially generalizable approach. Treatment of the human leukemia cell line ME-1 with these compounds shows decreased proliferation, indicating these are good candidates for further development.
- Published
- 2007
- Full Text
- View/download PDF
50. Structural Basis for Recognition of SMRT/N-CoR by the MYND Domain and Its Contribution to AML1/ETO's Activity
- Author
-
Justin J. Gaudet, Nancy A. Speck, Yizhou Liu, Wei Chen, Rachel C. Klet, Kari L. Hartman, Tomasz Cierpicki, John H. Bushweller, Liya Roudaia, Thomas M. Laue, and Matthew D. Cheney
- Subjects
Models, Molecular ,Cancer Research ,Oncogene Proteins, Fusion ,PROTEINS ,Cellular differentiation ,Gene Expression ,Repressor ,Bone Marrow Cells ,CELLCYCLE ,Plasma protein binding ,Biology ,medicine.disease_cause ,Article ,Cell Line ,Mice ,03 medical and health sciences ,RUNX1 Translocation Partner 1 Protein ,0302 clinical medicine ,Protein structure ,hemic and lymphatic diseases ,medicine ,Ring finger ,Animals ,Humans ,Nuclear Receptor Co-Repressor 1 ,neoplasms ,Nuclear receptor co-repressor 1 ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Mutation ,Nuclear Proteins ,Cell Differentiation ,Cell Biology ,Molecular biology ,Protein Structure, Tertiary ,3. Good health ,Repressor Proteins ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Core Binding Factor Alpha 2 Subunit ,Protein Binding - Abstract
SummaryAML1/ETO results from the t(8;21) associated with 12%–15% of acute myeloid leukemia. The AML1/ETO MYND domain mediates interactions with the corepressors SMRT and N-CoR and contributes to AML1/ETO's ability to repress proliferation and differentiation of primary bone marrow cells as well as to enhance their self renewal in vitro. We solved the solution structure of the MYND domain and show it to be structurally homologous to the PHD and RING finger families of proteins. We also determined the solution structure of an MYND-SMRT peptide complex. We demonstrated that a single amino acid substitution that disrupts the interaction between the MYND domain and the SMRT peptide attenuated AML1/ETO's effects on proliferation, differentiation, and gene expression.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.