35 results on '"Zhang, Yuannyu"'
Search Results
2. Metabolon formation regulates branched-chain amino acid oxidation and homeostasis
- Author
-
Patrick, McKenzie, Gu, Zhimin, Zhang, Gen, Wynn, R. Max, Kaphle, Pranita, Cao, Hui, Vu, Hieu, Cai, Feng, Gao, Xiaofei, Zhang, Yuannyu, Chen, Mingyi, Ni, Min, Chuang, David T., DeBerardinis, Ralph J., and Xu, Jian
- Published
- 2022
- Full Text
- View/download PDF
3. Structural variation cooperates with permissive chromatin to control enhancer hijacking–mediated oncogenic transcription
- Author
-
Botten, Giovanni A., Zhang, Yuannyu, Dudnyk, Kseniia, Kim, Yoon Jung, Liu, Xin, Sanders, Jacob T., Imanci, Aygun, Droin, Nathalie, Cao, Hui, Kaphle, Pranita, Dickerson, Kathryn E., Kumar, Kirthi R., Chen, Mingyi, Chen, Weina, Solary, Eric, Ly, Peter, Zhou, Jian, and Xu, Jian
- Published
- 2023
- Full Text
- View/download PDF
4. Epo-IGF1R cross talk expands stress-specific progenitors in regenerative erythropoiesis and myeloproliferative neoplasm
- Author
-
Hsieh, Hsi-Hsien, Yao, Huiyu, Ma, Yue, Zhang, Yuannyu, Xiao, Xue, Stephens, Helen, Wajahat, Naureen, Chung, Stephen S., Xu, Lin, Xu, Jian, Rampal, Raajit K., and Huang, Lily Jun-shen
- Published
- 2022
- Full Text
- View/download PDF
5. Regulation of mitochondrial biogenesis in erythropoiesis by mTORC1-mediated protein translation
- Author
-
Liu, Xin, Zhang, Yuannyu, Ni, Min, Cao, Hui, Signer, Robert AJ, Li, Dan, Li, Mushan, Gu, Zhimin, Hu, Zeping, Dickerson, Kathryn E, Weinberg, Samuel E, Chandel, Navdeep S, DeBerardinis, Ralph J, Zhou, Feng, Shao, Zhen, and Xu, Jian
- Subjects
Human Genome ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Genetics ,Stem Cell Research - Nonembryonic - Human ,Hematology ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Acetylation ,Animals ,Cells ,Cultured ,DNA-Binding Proteins ,Erythropoiesis ,Gene Expression Profiling ,Hematopoietic Stem Cells ,High Mobility Group Proteins ,Histones ,Humans ,Mechanistic Target of Rapamycin Complex 1 ,Mice ,Knockout ,Mitochondria ,Mitochondrial Proteins ,Multiprotein Complexes ,Organelle Biogenesis ,PTEN Phosphohydrolase ,Phenotype ,Prohibitins ,Protein Biosynthesis ,Proteomics ,RNA ,RNA Interference ,RNA ,Messenger ,RNA ,Mitochondrial ,Receptors ,Erythropoietin ,Repressor Proteins ,Signal Transduction ,TOR Serine-Threonine Kinases ,Transcription Factors ,Transfection ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Advances in genomic profiling present new challenges of explaining how changes in DNA and RNA are translated into proteins linking genotype to phenotype. Here we compare the genome-scale proteomic and transcriptomic changes in human primary haematopoietic stem/progenitor cells and erythroid progenitors, and uncover pathways related to mitochondrial biogenesis enhanced through post-transcriptional regulation. Mitochondrial factors including TFAM and PHB2 are selectively regulated through protein translation during erythroid specification. Depletion of TFAM in erythroid cells alters intracellular metabolism, leading to elevated histone acetylation, deregulated gene expression, and defective mitochondria and erythropoiesis. Mechanistically, mTORC1 signalling is enhanced to promote translation of mitochondria-associated transcripts through TOP-like motifs. Genetic and pharmacological perturbation of mitochondria or mTORC1 specifically impairs erythropoiesis in vitro and in vivo. Our studies support a mechanism for post-transcriptional control of erythroid mitochondria and may have direct relevance to haematologic defects associated with mitochondrial diseases and ageing.
- Published
- 2017
6. Silencing of LINE-1 retrotransposons is a selective dependency of myeloid leukemia
- Author
-
Gu, Zhimin, Liu, Yuxuan, Zhang, Yuannyu, Cao, Hui, Lyu, Junhua, Wang, Xun, Wylie, Annika, Newkirk, Simon J., Jones, Amanda E., Lee, Jr., Michael, Botten, Giovanni A., Deng, Mi, Dickerson, Kathryn E., Zhang, Cheng Cheng, An, Wenfeng, Abrams, John M., and Xu, Jian
- Published
- 2021
- Full Text
- View/download PDF
7. T/myeloid mixed phenotype acute leukaemia harbouring TLX3::BCL11B with TLX3 activation.
- Author
-
Botten, Giovanni A., Zhang, Yuannyu, Fuda, Franklin, Koduru, Prasad, Weinberg, Olga K., Slone, Tamra L., Zheng, Ruifang, Dickerson, Kathryn E., Gagan, Jeffrey R., and Chen, Weina
- Subjects
- *
ACUTE leukemia , *EXTRAMEDULLARY diseases , *GENE enhancers , *NUCLEOTIDE sequencing , *PHENOTYPES - Abstract
Summary: T/myeloid mixed phenotype acute leukaemia (MPAL) is a rare aggressive acute leukaemia with poorly understood pathogenesis. Herein, we report two cases of T/myeloid MPAL harbouring BCL11B‐associated structural variants that activate TLX3 (TLX3::BCL11B‐TLX3‐activation) by genome sequencing and transcriptomic analyses. Both patients were young males with extramedullary involvement. Cooperative gene alterations characteristic of T/myeloid MPAL and T‐lymphoblastic leukaemia (T‐ALL) were detected. Both patients achieved initial remission following lineage‐matched ALL‐based therapy with one patient requiring a lineage‐switched myeloid‐based therapy. Our study is the first to demonstrate the clinicopathological and genomic features of TLX3::BCL11B‐TLX3‐activated T/myeloid MPAL and provide insights into leukaemogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Convergence of oncogenic cooperation at single-cell and single-gene levels drives leukemic transformation
- Author
-
Liu, Yuxuan, Gu, Zhimin, Cao, Hui, Kaphle, Pranita, Lyu, Junhua, Zhang, Yuannyu, Hu, Wenhuo, Chung, Stephen S., Dickerson, Kathryn E., and Xu, Jian
- Published
- 2021
- Full Text
- View/download PDF
9. A unified model of human hemoglobin switching through single-cell genome editing
- Author
-
Shen, Yong, Verboon, Jeffrey M., Zhang, Yuannyu, Liu, Nan, Kim, Yoon Jung, Marglous, Samantha, Nandakumar, Satish K., Voit, Richard A., Fiorini, Claudia, Ejaz, Ayesha, Basak, Anindita, Orkin, Stuart H., Xu, Jian, and Sankaran, Vijay G.
- Published
- 2021
- Full Text
- View/download PDF
10. 3122 – CD44-MEDIATED METABOLIC REWIRING IS A TARGETABLE LIABILITY OF IDH-MUTANT LEUKEMIA
- Author
-
Lyu, Junhua, Liu, Yuxuan, Liu, Ningning, Vu, Hieu, Cai, Feng, Cao, Hui, Kaphle, Pranita, Wu, Zheng, Botten, Giovanni, Zhang, Yuannyu, Ni, Min, Chung, Stephen, DeBerardinis, Ralph, and Xu, Jian
- Published
- 2024
- Full Text
- View/download PDF
11. Multiplexed capture of spatial configuration and temporal dynamics of locus-specific 3D chromatin by biotinylated dCas9
- Author
-
Liu, Xin, Chen, Yong, Zhang, Yuannyu, Liu, Yuxuan, Liu, Nan, Botten, Giovanni A., Cao, Hui, Orkin, Stuart H., Zhang, Michael Q., and Xu, Jian
- Published
- 2020
- Full Text
- View/download PDF
12. Interrogation of enhancer function by enhancer-targeting CRISPR epigenetic editing
- Author
-
Li, Kailong, Liu, Yuxuan, Cao, Hui, Zhang, Yuannyu, Gu, Zhimin, Liu, Xin, Yu, Andy, Kaphle, Pranita, Dickerson, Kathryn E., Ni, Min, and Xu, Jian
- Published
- 2020
- Full Text
- View/download PDF
13. MAP: model-based analysis of proteomic data to detect proteins with significant abundance changes
- Author
-
Li, Mushan, Tu, Shiqi, Li, Zijia, Tan, Fengxiang, Liu, Jian, Wang, Qian, Zhang, Yuannyu, Xu, Jian, Zhang, Yijing, Zhou, Feng, and Shao, Zhen
- Published
- 2019
- Full Text
- View/download PDF
14. The mitochondrial respiratory chain is essential for haematopoietic stem cell function
- Author
-
Ansó, Elena, Weinberg, Samuel E., Diebold, Lauren P., Thompson, Benjamin J., Malinge, Sébastien, Schumacker, Paul T., Liu, Xin, Zhang, Yuannyu, Shao, Zhen, Steadman, Mya, Marsh, Kelly M., Xu, Jian, Crispino, John D., and Chandel, Navdeep S.
- Published
- 2017
- Full Text
- View/download PDF
15. Regulation of embryonic haematopoietic multipotency by EZH1
- Author
-
Vo, Linda T., Kinney, Melissa A., Liu, Xin, Zhang, Yuannyu, Barragan, Jessica, Sousa, Patricia M., Jha, Deepak K., Han, Areum, Cesana, Marcella, Shao, Zhen, North, Trista E., Orkin, Stuart H., Doulatov, Sergei, Xu, Jian, and Daley, George Q.
- Subjects
Hematopoiesis -- Research ,Embryonic development -- Research ,Physiological research -- Research ,Biological control systems -- Research ,Environmental issues ,Science and technology ,Zoology and wildlife conservation - Abstract
Author(s): Linda T. Vo [1, 2, 3]; Melissa A. Kinney [1, 2]; Xin Liu [4]; Yuannyu Zhang [4, 5]; Jessica Barragan [1, 2]; Patricia M. Sousa [1, 2]; Deepak K. [...]
- Published
- 2018
- Full Text
- View/download PDF
16. Targeting Cell Adhesion Molecules in IDH Mutant Myeloid Malignances
- Author
-
Lyu, Junhua, Liu, Yuxuan, Zhang, Yuannyu, Cai, Feng, Botten, Giovanni A., Cao, Hui, Kaphle, Pranita, Deberardinis, Ralph J, and Xu, Jian
- Published
- 2022
- Full Text
- View/download PDF
17. Structural Variation Cooperates with Permissive Chromatin to Control Enhancer Hijacking-Mediated Oncogenic Transcription
- Author
-
Botten, Giovanni A., Zhang, Yuannyu, Dudnyk, Kseniia, Liu, Xin, Sanders, Jacob T., Imanci, Aygun, Droin, Nathalie, Cao, Hui, Kaphle, Pranita, Dickerson, Kathryn E., Chen, Mingyi, Chen, Weina, Solary, Eric, Ly, Peter, Zhou, Jian, and Xu, Jian
- Published
- 2022
- Full Text
- View/download PDF
18. 3089 – ELUCIDATING NEW MECHANISMS OF ACQUIRED RESISTANCE TO IDH INHIBITION BY SATURATION VARIANT SCREENING OF BASE-EDITED LEUKEMIA CELLS
- Author
-
Lyu, Junhua, Liu, Yuxuan, Gong, Lihu, Chen, Mingyi, Madanat, Yazan, Zhang, Yuannyu, Gu, Zhimin, Cai, Feng, Cao, Hui, Kaphle, Pranita, Dickerson, Kathryn, Chen, Weina, Chung, Stephen, Collins, Robert, DeBerardinis, Ralph, Liu, Xin, and Xu, Jian
- Published
- 2021
- Full Text
- View/download PDF
19. Targeting the Epigenetic and Metabolic Vulnerabilities of Ezh2-Deficiency-Induced Myeloid Neoplasms
- Author
-
Gu, Zhimin, Liu, Yuxuan, Cao, Hui, Chen, Mingyi, Zhang, Yuannyu, Qi, Le, Liu, Xin, Li, Kailong, Dickerson, Kathryn, Cai, Feng, Chen, Weina, Ni, Min, Deberardinis, Ralph, and Xu, Jian
- Published
- 2017
- Full Text
- View/download PDF
20. Dissecting super-enhancer hierarchy based on chromatin interactions
- Author
-
Huang, Jialiang, Li, Kailong, Cai, Wenqing, Liu, Xin, Zhang, Yuannyu, Orkin, Stuart H., Xu, Jian, and Yuan, Guo-Cheng
- Abstract
Recent studies have highlighted super-enhancers (SEs) as important regulatory elements for gene expression, but their intrinsic properties remain incompletely characterized. Through an integrative analysis of Hi-C and ChIP-seq data, here we find that a significant fraction of SEs are hierarchically organized, containing both hub and non-hub enhancers. Hub enhancers share similar histone marks with non-hub enhancers, but are distinctly associated with cohesin and CTCF binding sites and disease-associated genetic variants. Genetic ablation of hub enhancers results in profound defects in gene activation and local chromatin landscape. As such, hub enhancers are the major constituents responsible for SE functional and structural organization.
- Published
- 2018
- Full Text
- View/download PDF
21. Quantitative Proteomic and Transcriptomic Analysis Reveals Post-Transcriptional Regulation of Mitochondrial Biogenesis during Erythropoiesis
- Author
-
Liu, Xin, Zhang, Yuannyu, Zhou, Feng, Shao, Zhen, Signer, Robert, Cao, Hui, Hu, Zeping, DeBerardinis, Ralph, and Xu, Jian
- Published
- 2015
- Full Text
- View/download PDF
22. Lactate Dehydrogenase A Governs Cardiac Hypertrophic Growth in Response to Hemodynamic Stress.
- Author
-
Dai, Chongshan, Li, Qinfeng, May, Herman I., Li, Chao, Zhang, Guangyu, Sharma, Gaurav, Sherry, A. Dean, Malloy, Craig R., Khemtong, Chalermchai, Zhang, Yuannyu, Deng, Yingfeng, Gillette, Thomas G., Xu, Jian, Scadden, David T., and Wang, Zhao V.
- Abstract
The heart manifests hypertrophic growth in response to high blood pressure, which may decompensate and progress to heart failure under persistent stress. Metabolic remodeling is an early event in this process. However, its role remains to be fully characterized. Here, we show that lactate dehydrogenase A (LDHA), a critical glycolytic enzyme, is elevated in the heart in response to hemodynamic stress. Cardiomyocyte-restricted deletion of LDHA leads to defective cardiac hypertrophic growth and heart failure by pressure overload. Silencing of LDHA in cultured cardiomyocytes suppresses cell growth from pro-hypertrophic stimulation in vitro , while overexpression of LDHA is sufficient to drive cardiomyocyte growth. Furthermore, we find that lactate is capable of rescuing the growth defect from LDHA knockdown. Mechanistically, lactate stabilizes NDRG3 (N-myc downregulated gene family 3) and stimulates ERK (extracellular signal-regulated kinase). Our results together suggest that the LDHA/NDRG3 axis may play a critical role in adaptive cardiomyocyte growth in response to hemodynamic stress. • Metabolic remodeling plays an important role in hypertensive heart disease • LDHA as a key enzyme of glycolysis is increased in the hypertrophic heart • Deficiency of LDHA exacerbates cardiomyopathy under pressure overload • LDHA may be required for adaptive hypertrophic growth via NDRG3 stimulation Dai et al. find that LDHA is significantly increased in the heart under hemodynamic stress, and cardiomyocyte-specific deletion of LDHA leads to severe cardiac dysfunction in response to pressure overload. LDHA may govern adaptive growth through elevation of NDRG3 and activation of ERK. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
23. Discovering How Heme Controls Genome Function Through Heme-omics.
- Author
-
Liao, Ruiqi, Zheng, Ye, Liu, Xin, Zhang, Yuannyu, Seim, Gretchen, Tanimura, Nobuyuki, Wilson, Gary M., Hematti, Peiman, Coon, Joshua J., Fan, Jing, Xu, Jian, Keles, Sunduz, and Bresnick, Emery H.
- Abstract
Protein ensembles control genome function by establishing, maintaining, and deconstructing cell-type-specific chromosomal landscapes. A plethora of small molecules orchestrate cellular functions and therefore may link physiological processes with genome biology. The metabolic enzyme and hemoglobin cofactor heme induces proteolysis of a transcriptional repressor, Bach1, and regulates gene expression post-transcriptionally. However, whether heme controls genome function broadly or through prescriptive actions is unclear. Using assay for transposase-accessible chromatin sequencing (ATAC-seq), we establish a heme-dependent chromatin atlas in wild-type and mutant erythroblasts lacking enhancers that confer normal heme synthesis. Amalgamating chromatin landscapes and transcriptomes in cells with sub-physiological heme and post-heme rescue reveals parallel Bach1-dependent and Bach1-independent mechanisms that target heme-sensing chromosomal hotspots. The hotspots harbor a DNA motif demarcating heme-regulated chromatin and genes encoding proteins not known to be heme regulated, including metabolic enzymes. The heme-omics analysis establishes how an essential biochemical cofactor controls genome function and cellular physiology. • Heme-omics resource is generated by amalgamating ATAC-seq and RNA-seq datasets • Parallel Bach1-dependent and independent heme mechanisms regulate genome function • A unique DNA motif demarcates heme-regulated chromatin sites • Heme-sensing hotspots reveal new dimensions in genome biology and cellular regulation Liao et al. generate a heme-regulated chromatin atlas by amalgamating ATAC-seq and RNA-seq datasets from cells with normal and sub-physiological heme, and they identify parallel Bach1-dependent and Bach1-independent mechanisms that target heme-sensing chromatin hotspots. The hotspots harbor a DNA motif demarcating heme-regulated chromatin and genes not known to be heme regulated. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
24. In Situ Capture of Chromatin Interactions by Biotinylated dCas9.
- Author
-
Liu, Xin, Zhang, Yuannyu, Chen, Yong, Li, Mushan, Zhou, Feng, Li, Kailong, Cao, Hui, Ni, Min, Liu, Yuxuan, Gu, Zhimin, Dickerson, Kathryn E., Xie, Shiqi, Hon, Gary C., Xuan, Zhenyu, Zhang, Michael Q., Shao, Zhen, and Xu, Jian
- Subjects
- *
CHROMATIN , *CRISPRS , *LOCUS (Genetics) , *NUCLEASES , *TELOMERES - Abstract
Summary Cis -regulatory elements (CREs) are commonly recognized by correlative chromatin features, yet the molecular composition of the vast majority of CREs in chromatin remains unknown. Here, we describe a CRISPR affinity purification in situ of regulatory elements (CAPTURE) approach to unbiasedly identify locus-specific chromatin-regulating protein complexes and long-range DNA interactions. Using an in vivo biotinylated nuclease-deficient Cas9 protein and sequence-specific guide RNAs, we show high-resolution and selective isolation of chromatin interactions at a single-copy genomic locus. Purification of human telomeres using CAPTURE identifies known and new telomeric factors. In situ capture of individual constituents of the enhancer cluster controlling human β-globin genes establishes evidence for composition-based hierarchical organization. Furthermore, unbiased analysis of chromatin interactions at disease-associated cis -elements and developmentally regulated super-enhancers reveals spatial features that causally control gene transcription. Thus, comprehensive and unbiased analysis of locus-specific regulatory composition provides mechanistic insight into genome structure and function in development and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
25. Dynamic Control of Enhancer Repertoires Drives Lineage and Stage-Specific Transcription during Hematopoiesis.
- Author
-
Huang, Jialiang, Liu, Xin, Li, Dan, Shao, Zhen, Cao, Hui, Zhang, Yuannyu, Trompouki, Eirini, Bowman, Teresa V., Zon, Leonard I., Yuan, Guo-Cheng, Orkin, Stuart H., and Xu, Jian
- Subjects
- *
GENETIC transcription , *HEMATOPOIESIS , *GENETIC regulation , *HEMATOPOIETIC stem cells , *PURE red cell aplasia - Abstract
Summary Enhancers are the primary determinants of cell identity, but the regulatory components controlling enhancer turnover during lineage commitment remain largely unknown. Here we compare the enhancer landscape, transcriptional factor occupancy, and transcriptomic changes in human fetal and adult hematopoietic stem/progenitor cells and committed erythroid progenitors. We find that enhancers are modulated pervasively and direct lineage- and stage-specific transcription. GATA2-to-GATA1 switch is prevalent at dynamic enhancers and drives erythroid enhancer commissioning. Examination of lineage-specific enhancers identifies transcription factors and their combinatorial patterns in enhancer turnover. Importantly, by CRISPR/Cas9-mediated genomic editing, we uncover functional hierarchy of constituent enhancers within the SLC25A37 super-enhancer. Despite indistinguishable chromatin features, we reveal through genomic editing the functional diversity of several GATA switch enhancers in which enhancers with opposing functions cooperate to coordinate transcription. Thus, genome-wide enhancer profiling coupled with in situ enhancer editing provide critical insights into the functional complexity of enhancers during development. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
26. GATA/Heme Multi-omics Reveals a Trace Metal-Dependent Cellular Differentiation Mechanism.
- Author
-
Tanimura, Nobuyuki, Liao, Ruiqi, Wilson, Gary M., Dent, Matthew R., Cao, Miao, Burstyn, Judith N., Hematti, Peiman, Liu, Xin, Zhang, Yuannyu, Zheng, Ye, Keles, Sunduz, Xu, Jian, Coon, Joshua J., and Bresnick, Emery H.
- Subjects
- *
HEME , *ENZYMES , *HEMOGLOBINS , *ERYTHROCYTES , *THALASSEMIA - Abstract
Summary By functioning as an enzyme cofactor, hemoglobin component, and gene regulator, heme is vital for life. One mode of heme-regulated transcription involves amplifying the activity of GATA-1, a key determinant of erythrocyte differentiation. To discover biological consequences of the metal cofactor-transcription factor mechanism, we merged GATA-1/heme-regulated sectors of the proteome and transcriptome. This multi-omic analysis revealed a GATA-1/heme circuit involving hemoglobin subunits, ubiquitination components, and proteins not implicated in erythrocyte biology, including the zinc exporter Slc30a1. Though GATA-1 induced expression of Slc30a1 and the zinc importer Slc39a8, Slc39a8 dominantly increased intracellular zinc, which conferred erythroblast survival. Subsequently, a zinc transporter switch, involving decreased importer and sustained exporter expression, reduced intracellular zinc during terminal differentiation. Downregulating Slc30a1 increased intracellular zinc and, strikingly, accelerated differentiation. This analysis established a conserved paradigm in which a GATA-1/heme circuit controls trace metal transport machinery and trace metal levels as a mechanism governing cellular differentiation. Graphical Abstract Highlights • Multi-omic analysis yields resource for exploring erythrocyte development regulation • GATA-1/heme induce a zinc transporter switch that controls intracellular zinc levels • Zinc confers erythroblast survival • Decreased zinc restricts and increased zinc promotes terminal differentiation Zinc deficiency causes anemia through poorly understood mechanisms. Tanimura et al. report that GATA-1, a major determinant of red blood cell development, and heme, an essential cofactor for hemoglobin synthesis, control zinc levels. Zinc levels in turn regulate red blood cell development, thereby establishing a paradigm that informs anemia mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
27. A glutamine metabolic switch supports erythropoiesis.
- Author
-
Lyu J, Gu Z, Zhang Y, Vu HS, Lechauve C, Cai F, Cao H, Keith J, Brancaleoni V, Granata F, Motta I, Cappellini MD, Huang LJ, DeBerardinis RJ, Weiss MJ, Ni M, and Xu J
- Subjects
- Animals, Humans, Mice, Erythroid Precursor Cells metabolism, Glutamic Acid metabolism, Heme metabolism, Oxidation-Reduction, Mice, Knockout, Ammonium Compounds metabolism, beta-Thalassemia metabolism, beta-Thalassemia genetics, Erythropoiesis, Glutamate-Ammonia Ligase metabolism, Glutamate-Ammonia Ligase genetics, Glutamine metabolism, Oxidative Stress
- Abstract
Metabolic requirements vary during development, and our understanding of how metabolic activity influences cell specialization is incomplete. Here, we describe a switch from glutamine catabolism to synthesis required for erythroid cell maturation. Glutamine synthetase (GS), one of the oldest functioning genes in evolution, is activated during erythroid maturation to detoxify ammonium generated from heme biosynthesis, which is up-regulated to support hemoglobin production. Loss of GS in mouse erythroid precursors caused ammonium accumulation and oxidative stress, impairing erythroid maturation and recovery from anemia. In β-thalassemia, GS activity is inhibited by protein oxidation, leading to glutamate and ammonium accumulation, whereas enhancing GS activity alleviates the metabolic and pathological defects. Our findings identify an evolutionarily conserved metabolic adaptation that could potentially be leveraged to treat common red blood cell disorders.
- Published
- 2024
- Full Text
- View/download PDF
28. Electron transport chain inhibition increases cellular dependence on purine transport and salvage.
- Author
-
Wu Z, Bezwada D, Harris RC, Pan C, Nguyen PT, Faubert B, Cai L, Cai F, Vu HS, Chen H, Sandoval MM, Do D, Gu W, Zhang Y, Ko B, Brooks B, Kelekar S, Zhang Y, Zacharias LG, Oaxaca KC, Mathews TP, Garcia-Bermudez J, Ni M, and DeBerardinis RJ
- Abstract
Cancer cells reprogram their metabolism to support cell growth and proliferation in harsh environments. While many studies have documented the importance of mitochondrial oxidative phosphorylation (OXPHOS) in tumor growth, some cancer cells experience conditions of reduced OXPHOS in vivo and induce alternative metabolic pathways to compensate. To assess how human cells respond to mitochondrial dysfunction, we performed metabolomics in fibroblasts and plasma from patients with inborn errors of mitochondrial metabolism, and in cancer cells subjected to inhibition of the electron transport chain (ETC). All these analyses revealed extensive perturbations in purine-related metabolites; in non-small cell lung cancer (NSCLC) cells, ETC blockade led to purine metabolite accumulation arising from a reduced cytosolic NAD
+ /NADH ratio (NADH reductive stress). Stable isotope tracing demonstrated that ETC deficiency suppressed de novo purine nucleotide synthesis while enhancing purine salvage. Analysis of NSCLC patients infused with [U-13 C]glucose revealed that tumors with markers of low oxidative mitochondrial metabolism exhibited high expression of the purine salvage enzyme HPRT1 and abundant levels of the HPRT1 product inosine monophosphate (IMP). ETC blockade also induced production of ribose-5' phosphate (R5P) by the pentose phosphate pathway (PPP) and import of purine nucleobases. Blocking either HPRT1 or nucleoside transporters sensitized cancer cells to ETC inhibition, and overexpressing nucleoside transporters was sufficient to drive growth of NSCLC xenografts. Collectively, this study mechanistically delineates how cells compensate for suppressed purine metabolism in response to ETC blockade, and uncovers a new metabolic vulnerability in tumors experiencing NADH excess.- Published
- 2023
- Full Text
- View/download PDF
29. Light-activated macromolecular phase separation modulates transcription by reconfiguring chromatin interactions.
- Author
-
Kim YJ, Lee M Jr, Lee YT, Jing J, Sanders JT, Botten GA, He L, Lyu J, Zhang Y, Mettlen M, Ly P, Zhou Y, and Xu J
- Subjects
- Cell Nucleus genetics, Transcription Factors genetics, Genome, Chromatin genetics, Proteomics
- Abstract
Biomolecular condensates participate in the regulation of gene transcription, yet the relationship between nuclear condensation and transcriptional activation remains elusive. Here, we devised a biotinylated CRISPR-dCas9-based optogenetic method, light-activated macromolecular phase separation (LAMPS), to enable inducible formation, affinity purification, and multiomic dissection of nuclear condensates at the targeted genomic loci. LAMPS-induced condensation at enhancers and promoters activates endogenous gene transcription by chromatin reconfiguration, causing increased chromatin accessibility and de novo formation of long-range chromosomal loops. Proteomic profiling of light-induced condensates by dCas9-mediated affinity purification uncovers multivalent interaction-dependent remodeling of macromolecular composition, resulting in the selective enrichment of transcriptional coactivators and chromatin structure proteins. Our findings support a model whereby the formation of nuclear condensates at native genomic loci reconfigures chromatin architecture and multiprotein assemblies to modulate gene transcription. Hence, LAMPS facilitates mechanistic interrogation of the relationship between nuclear condensation, genome structure, and gene transcription in living cells.
- Published
- 2023
- Full Text
- View/download PDF
30. Disabling Uncompetitive Inhibition of Oncogenic IDH Mutations Drives Acquired Resistance.
- Author
-
Lyu J, Liu Y, Gong L, Chen M, Madanat YF, Zhang Y, Cai F, Gu Z, Cao H, Kaphle P, Kim YJ, Kalkan FN, Stephens H, Dickerson KE, Ni M, Chen W, Patel P, Mims AS, Borate U, Burd A, Cai SF, Yin CC, You MJ, Chung SS, Collins RH, DeBerardinis RJ, Liu X, and Xu J
- Subjects
- Humans, NADP, Mutation, Amino Acids genetics, Isocitrate Dehydrogenase, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism
- Abstract
Mutations in IDH genes occur frequently in acute myeloid leukemia (AML) and other human cancers to generate the oncometabolite R-2HG. Allosteric inhibition of mutant IDH suppresses R-2HG production in a subset of patients with AML; however, acquired resistance emerges as a new challenge, and the underlying mechanisms remain incompletely understood. Here we establish isogenic leukemia cells containing common IDH oncogenic mutations by CRISPR base editing. By mutational scanning of IDH single amino acid variants in base-edited cells, we describe a repertoire of IDH second-site mutations responsible for therapy resistance through disabling uncompetitive enzyme inhibition. Recurrent mutations at NADPH binding sites within IDH heterodimers act in cis or trans to prevent the formation of stable enzyme-inhibitor complexes, restore R-2HG production in the presence of inhibitors, and drive therapy resistance in IDH-mutant AML cells and patients. We therefore uncover a new class of pathogenic mutations and mechanisms for acquired resistance to targeted cancer therapies., Significance: Comprehensive scanning of IDH single amino acid variants in base-edited leukemia cells uncovers recurrent mutations conferring resistance to IDH inhibition through disabling NADPH-dependent uncompetitive inhibition. Together with targeted sequencing, structural, and functional studies, we identify a new class of pathogenic mutations and mechanisms for acquired resistance to IDH-targeting cancer therapies. This article is highlighted in the In This Issue feature, p. 1., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
31. Noncoding Variants Connect Enhancer Dysregulation with Nuclear Receptor Signaling in Hematopoietic Malignancies.
- Author
-
Li K, Zhang Y, Liu X, Liu Y, Gu Z, Cao H, Dickerson KE, Chen M, Chen W, Shao Z, Ni M, and Xu J
- Subjects
- Humans, Hematologic Neoplasms genetics, Receptors, Cytoplasmic and Nuclear metabolism
- Abstract
Mutations in protein-coding genes are well established as the basis for human cancer, yet how alterations within noncoding genome, a substantial fraction of which contain cis -regulatory elements (CRE), contribute to cancer pathophysiology remains elusive. Here, we developed an integrative approach to systematically identify and characterize noncoding regulatory variants with functional consequences in human hematopoietic malignancies. Combining targeted resequencing of hematopoietic lineage-associated CREs and mutation discovery, we uncovered 1,836 recurrently mutated CREs containing leukemia-associated noncoding variants. By enhanced CRISPR/dCas9-based CRE perturbation screening and functional analyses, we identified 218 variant-associated oncogenic or tumor-suppressive CREs in human leukemia. Noncoding variants at KRAS and PER2 enhancers reside in proximity to nuclear receptor (NR) binding regions and modulate transcriptional activities in response to NR signaling in leukemia cells. NR binding sites frequently colocalize with noncoding variants across cancer types. Hence, recurrent noncoding variants connect enhancer dysregulation with nuclear receptor signaling in hematopoietic malignancies. SIGNIFICANCE: We describe an integrative approach to identify noncoding variants in human leukemia, and reveal cohorts of variant-associated oncogenic and tumor-suppressive cis -regulatory elements including KRAS and PER2 enhancers. Our findings support a model in which noncoding regulatory variants connect enhancer dysregulation with nuclear receptor signaling to modulate gene programs in hematopoietic malignancies. See related commentary by van Galen, p. 646 . This article is highlighted in the In This Issue feature, p. 627 ., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
32. Loss of EZH2 Reprograms BCAA Metabolism to Drive Leukemic Transformation.
- Author
-
Gu Z, Liu Y, Cai F, Patrick M, Zmajkovic J, Cao H, Zhang Y, Tasdogan A, Chen M, Qi L, Liu X, Li K, Lyu J, Dickerson KE, Chen W, Ni M, Merritt ME, Morrison SJ, Skoda RC, DeBerardinis RJ, and Xu J
- Subjects
- Amino Acids, Branched-Chain metabolism, Animals, Enhancer of Zeste Homolog 2 Protein metabolism, Humans, Leukemia genetics, Leukemia metabolism, Mice, Mutation, Myeloproliferative Disorders complications, Myeloproliferative Disorders metabolism, Neoplasm Transplantation, Signal Transduction, TOR Serine-Threonine Kinases metabolism, Enhancer of Zeste Homolog 2 Protein genetics, GTP Phosphohydrolases genetics, Leukemia pathology, Membrane Proteins genetics, Myeloproliferative Disorders genetics, Transaminases metabolism
- Abstract
Epigenetic gene regulation and metabolism are highly intertwined, yet little is known about whether altered epigenetics influence cellular metabolism during cancer progression. Here, we show that EZH2 and NRAS
G12D mutations cooperatively induce progression of myeloproliferative neoplasms to highly penetrant, transplantable, and lethal myeloid leukemias in mice. EZH1, an EZH2 homolog, is indispensable for EZH2-deficient leukemia-initiating cells and constitutes an epigenetic vulnerability. BCAT1, which catalyzes the reversible transamination of branched-chain amino acids (BCAA), is repressed by EZH2 in normal hematopoiesis and aberrantly activated in EZH2-deficient myeloid neoplasms in mice and humans. BCAT1 reactivation cooperates with NRASG12D to sustain intracellular BCAA pools, resulting in enhanced mTOR signaling in EZH2-deficient leukemia cells. Genetic and pharmacologic inhibition of BCAT1 selectively impairs EZH2-deficient leukemia-initiating cells and constitutes a metabolic vulnerability. Hence, epigenetic alterations rewire intracellular metabolism during leukemic transformation, causing epigenetic and metabolic vulnerabilities in cancer-initiating cells. SIGNIFICANCE: EZH2 inactivation and oncogenic NRAS cooperate to induce leukemic transformation of myeloproliferative neoplasms by activating BCAT1 to enhance BCAA metabolism and mTOR signaling. We uncover a mechanism by which epigenetic alterations rewire metabolism during cancer progression, causing epigenetic and metabolic liabilities in cancer-initiating cells that may be exploited as potential therapeutics. See related commentary by Li and Melnick, p. 1158 . This article is highlighted in the In This Issue feature, p. 1143 ., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
33. Installation of a cancer promoting WNT/SIX1 signaling axis by the oncofusion protein MLL-AF9.
- Author
-
Zhang LS, Kang X, Lu J, Zhang Y, Wu X, Wu G, Zheng J, Tuladhar R, Shi H, Wang Q, Morlock L, Yao H, Huang LJ, Maire P, Kim J, Williams N, Xu J, Chen C, Zhang CC, and Lum L
- Subjects
- Animals, HEK293 Cells, HL-60 Cells, HeLa Cells, Humans, Intracellular Signaling Peptides and Proteins genetics, Leukemia, Myeloid, Acute metabolism, Mice, Neoplasm Transplantation, Neoplastic Stem Cells metabolism, Receptors, G-Protein-Coupled genetics, THP-1 Cells, Transcription Factor 7-Like 2 Protein metabolism, Homeodomain Proteins genetics, Leukemia, Myeloid, Acute genetics, Myeloid-Lymphoid Leukemia Protein genetics, Oncogene Proteins, Fusion genetics, Small Molecule Libraries pharmacology, Wnt Signaling Pathway drug effects
- Abstract
Background: Chromosomal translocation-induced expression of the chromatin modifying oncofusion protein MLL-AF9 promotes acute myelocytic leukemia (AML). Whereas WNT/β-catenin signaling has previously been shown to support MLL-AF9-driven leukemogenesis, the mechanism underlying this relationship remains unclear., Methods: We used two novel small molecules targeting WNT signaling as well as a genetically modified mouse model that allow targeted deletion of the WNT protein chaperone Wntless (WLS) to evaluate the role of WNT signaling in AML progression. ATAC-seq and transcriptome profiling were deployed to understand the cellular consequences of disrupting a WNT signaling in leukemic initiating cells (LICs)., Findings: We identified Six1 to be a WNT-controlled target gene in MLL-AF9-transformed leukemic initiating cells (LICs). MLL-AF9 alters the accessibility of Six1 DNA to the transcriptional effector TCF7L2, a transducer of WNT/β-catenin gene expression changes. Disruption of WNT/SIX1 signaling using inhibitors of the Wnt signaling delays the development of AML., Interpretation: By rendering TCF/LEF-binding elements controlling Six1 accessible to TCF7L2, MLL-AF9 promotes WNT/β-catenin-dependent growth of LICs. Small molecules disrupting WNT/β-catenin signaling block Six1 expression thereby disrupting leukemia driven by MLL fusion proteins., (Copyright © 2018 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
34. CAPTURE: In Situ Analysis of Chromatin Composition of Endogenous Genomic Loci by Biotinylated dCas9.
- Author
-
Liu X, Zhang Y, Chen Y, Li M, Shao Z, Zhang MQ, and Xu J
- Subjects
- Cell Line, Humans, Regulatory Elements, Transcriptional, CRISPR-Associated Protein 9 chemistry, Chromatin chemistry, Clustered Regularly Interspaced Short Palindromic Repeats, Genomics methods
- Abstract
Cis-regulatory elements (CREs) play a pivotal role in spatiotemporal control of tissue-specific gene expression, yet the molecular composition of the vast majority of CREs in native chromatin remains unknown. In this article, we describe the clustered regularly interspaced short palindromic repeats (CRISPR) affinity purification in situ of regulatory elements (CAPTURE) approach to simultaneously identify locus-specific chromatin-regulating protein complexes and long-range DNA interactions. Using an in vivo biotinylated nuclease-deficient Cas9 (dCas9) protein and programmable single guide RNAs (sgRNAs), this approach allows for high-resolution and locus-specific isolation of protein complexes and long-range chromatin looping associated with single copy CREs in mammalian cells. Unbiased analysis of the compositional structure of developmentally regulated or disease-associated CREs identifies new features of transcriptional regulation. Hence, CAPTURE provides a versatile platform to study genomic locus-regulating chromatin composition in a mammalian genome. © 2018 by John Wiley & Sons, Inc., (© 2018 John Wiley & Sons, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
35. Temporal dynamics of cardiac hypertrophic growth in response to pressure overload.
- Author
-
Wang Y, Zhang Y, Ding G, May HI, Xu J, Gillette TG, Wang H, and Wang ZV
- Subjects
- Animals, Animals, Newborn, Aorta, Thoracic surgery, Cardiomegaly etiology, Cardiomegaly metabolism, Cardiomegaly physiopathology, Cells, Cultured, Constriction, Disease Models, Animal, Gene Expression Regulation, Male, Mice, Inbred C57BL, Muscle Proteins biosynthesis, Muscle Proteins genetics, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Protein Biosynthesis, Puromycin metabolism, Rats, Sprague-Dawley, Time Factors, Aorta, Thoracic physiopathology, Arterial Pressure, Cardiomegaly pathology, Cell Proliferation drug effects, Myocytes, Cardiac pathology
- Abstract
Hypertension is one of the most important risk factors of heart failure. In response to high blood pressure, the left ventricle manifests hypertrophic growth to ameliorate wall stress, which may progress into decompensation and trigger pathological cardiac remodeling. Despite the clinical importance, the temporal dynamics of pathological cardiac growth remain elusive. Here, we took advantage of the puromycin labeling approach to measure the relative rates of protein synthesis as a way to delineate the temporal regulation of cardiac hypertrophic growth. We first identified the optimal treatment conditions for puromycin in neonatal rat ventricular myocyte culture. We went on to demonstrate that myocyte growth reached its peak rate after 8-10 h of growth stimulation. At the in vivo level, with the use of an acute surgical model of pressure-overload stress, we observed the maximal growth rate to occur at day 7 after surgery. Moreover, RNA sequencing analysis supports that the most profound transcriptomic changes occur during the early phase of hypertrophic growth. Our results therefore suggest that cardiac myocytes mount an immediate growth response in reply to pressure overload followed by a gradual return to basal levels of protein synthesis, highlighting the temporal dynamics of pathological cardiac hypertrophic growth. NEW & NOTEWORTHY We determined the optimal conditions of puromycin incorporation in cardiac myocyte culture. We took advantage of this approach to identify the growth dynamics of cardiac myocytes in vitro. We went further to discover the protein synthesis rate in vivo, which provides novel insights about cardiac temporal growth dynamics in response to pressure overload., (Copyright © 2017 the American Physiological Society.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.