34 results on '"Xiaoyang Su"'
Search Results
2. Mitochondrial uncoupler MB1-47 is efficacious in treating hepatic metastasis of pancreatic cancer in murine tumor transplantation models
- Author
-
Bin Cao, Shengkan Jin, Victor M. Tan, Amer Alasadi, Jingjing Guo, Juan Collantes, Hanlin Tao, Xiaoyang Su, and David Augeri
- Subjects
0301 basic medicine ,Cancer Research ,Citric Acid Cycle ,Adenocarcinoma ,Mitochondrion ,Biology ,Mice ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,Cell Line, Tumor ,Pancreatic cancer ,Pyruvic Acid ,Genetics ,medicine ,Animals ,Humans ,Glycolysis ,Molecular Biology ,Cell Proliferation ,Futile cycle ,Liver Neoplasms ,Cell Cycle Checkpoints ,Cell cycle ,medicine.disease ,Warburg effect ,Adenosine Monophosphate ,Mitochondria ,Adenosine Diphosphate ,Transplantation ,Disease Models, Animal ,Glucose ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Heterografts ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDA) is aggressive cancer characterized by rapid progression, metastatic recurrence, and highly resistant to treatment. PDA cells exhibit aerobic glycolysis, or the Warburg effect, which reduces the flux of pyruvate into mitochondria. As a result, more glycolytic metabolites are shunted to pathways for the production of building blocks (e.g., ribose) and reducing agents (e.g., NADPH) for biosynthesis that are necessary for cell proliferation. In addition, PDA cells are highly addicted to glutamine for both maintaining biosynthetic pathways and achieving redox balance. Mitochondrial uncoupling facilitates proton influx across the mitochondrial inner membrane without generating ATP, leading to a futile cycle that consumes glucose metabolites and glutamine. We synthesized a new mitochondrial uncoupler MB1-47 and tested its effect on cancer cell metabolism and the anticancer activity in pancreatic cancer cell models and murine tumor transplantation models. MB1-47 uncouples mitochondria in the pancreatic cancer cells, resulting in: (1) the acceleration of pyruvate oxidation and TCA turnover; (2) increases in AMP/ATP and ADP/AMP ratios; and (3) a decrease in the synthesis rate of nucleotides and sugar nucleotides. Moreover, MB1-47 arrests cell cycle at G0-G1 phase, reduces clonogenicity, and inhibits cell growth of murine and human pancreatic cancer cells. In vivo studies showed that MB1-47 inhibits tumor growth in murine tumor transplantation models, and inhibits the hepatic metastasis when tumor cells were transplanted intrasplenically. Our results provide proof of concept for a potentially new strategy of treating PDA, and a novel prototype experimental drug for future studies and development.
- Published
- 2021
3. A therapeutically targetable NOTCH1-SIRT1-KAT7 axis in T-cell Leukemia
- Author
-
Olga Lancho, Amartya Singh, Victoria da Silva-Diz, Maya Aleksandrova, Jesminara Khatun, Luca Tottone, Patricia Renck Nunes, Shirley Luo, Caifeng Zhao, Haiyan Zheng, Eric Chiles, Zhenyu Zuo, Pedro P. Rocha, Xiaoyang Su, Hossein Khiabanian, and Daniel Herranz
- Subjects
Leukemia, T-Cell ,Sirtuin 1 ,Acetyltransferases ,Humans ,General Medicine ,Receptor, Notch1 ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Research Articles ,Signal Transduction ,Histone Acetyltransferases - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a NOTCH1-driven disease in need of novel therapies. Here, we identify a NOTCH1–SIRT1–KAT7 link as a therapeutic vulnerability in T-ALL, in which the histone deacetylase SIRT1 is overexpressed downstream of a NOTCH1-bound enhancer. SIRT1 loss impaired leukemia generation, whereas SIRT1 overexpression accelerated leukemia and conferred resistance to NOTCH1 inhibition in a deacetylase-dependent manner. Moreover, pharmacologic or genetic inhibition of SIRT1 resulted in significant antileukemic effects. Global acetyl proteomics upon SIRT1 loss uncovered hyperacetylation of KAT7 and BRD1, subunits of a histone acetyltransferase complex targeting H4K12. Metabolic and gene-expression profiling revealed metabolic changes together with a transcriptional signature resembling KAT7 deletion. Consistently, SIRT1 loss resulted in reduced H4K12ac, and overexpression of a nonacetylatable KAT7-mutant partly rescued SIRT1 loss-induced proliferation defects. Overall, our results uncover therapeutic targets in T-ALL and reveal a circular feedback mechanism balancing deacetylase/acetyltransferase activation with potentially broad relevance in cancer. Significance: We identify a T-ALL axis whereby NOTCH1 activates SIRT1 through an enhancer region, and SIRT1 deacetylates and activates KAT7. Targeting SIRT1 shows antileukemic effects, partly mediated by KAT7 inactivation. Our results reveal T-ALL therapeutic targets and uncover a rheostat mechanism between deacetylase/acetyltransferase activities with potentially broader cancer relevance. This article is highlighted in the In This Issue feature, p. 1
- Published
- 2022
4. Butyrate Drives Metabolic Rewiring and Epigenetic Reprogramming in Human Colon Cancer Cells
- Author
-
Lujing Wang, Ahmad Abdel Fat Shannar, Renyi Wu, Pochung Chou, Md Shahid Sarwar, Hsiao‐chen Kuo, Rebecca Mary Peter, Yujue Wang, Xiaoyang Su, and Ah‐Ng Kong
- Subjects
Epigenomics ,Histone Demethylases ,Kelch-Like ECH-Associated Protein 1 ,NF-E2-Related Factor 2 ,DNA Methylation ,Article ,Epigenesis, Genetic ,Mixed Function Oxygenases ,Proto-Oncogene Proteins ,Colonic Neoplasms ,Butyric Acid ,Humans ,Food Science ,Biotechnology - Abstract
SCOPE: Butyrate (B) is a short-chain fatty acid produced by dietary fiber, known to inhibit histone deacetylases (HDACs) and possess cancer-preventive/anti-cancer effects. However, the role of B in metabolic rewiring, epigenomic reprogramming, transcriptomic network, NRF2 signaling and eliciting cancer-preventive effects in colorectal cancer (CRC) HCT116 cell remains unclear. METHODS AND RESULTS: Sodium butyrate (NaB) dose-dependently inhibited the growth of CRC HCT116 cells. NaB inhibited NRF2/NRF2-target genes and blocked NRF2-ARE signaling. NaB increased NRF2 negative regulator KEAP1 expression through inhibiting its promoter methylation. Associative analysis of DEGs (differentially expressed genes) from RNA-seq and DMRs (differentially methylated regions) from CpG methyl-seq identified the tumor suppressor gene ABCA1 and tumor promote gene EGR3 were correlated with their promoters’ CpG methylation indicating NaB regulates cancer markers through modulating their promoter methylation. NaB activated the mitochondrial tricarboxylic acid (TCA) cycle while inhibited the methionine metabolism which are both tightly coupled to the epigenetic machinery. NaB regulated the epigenetic enzymes/genes including DNMT1, HAT1, KDM1A, KDM1B and TET1. Altogether, B’s regulation of metabolites coupled to the epigenetic enzymes illustrates the potential underlying biological connectivity between metabolomics and epigenomics. CONCLUSION: B regulates KEAP1/NRF2 signaling, drives metabolic rewiring, CpG methylomic and transcriptomic reprogramming contributing to the overall cancer-prevention/anti-cancer effect in the CRC cell model.
- Published
- 2022
5. Development of a portable toolkit to diagnose coral thermal stress
- Author
-
Zhuolun Meng, Amanda Williams, Pinky Liau, Timothy G. Stephens, Crawford Drury, Eric N. Chiles, Xiaoyang Su, Mehdi Javanmard, and Debashish Bhattacharya
- Subjects
Multidisciplinary ,Coral Reefs ,Animals ,Humans ,Anthozoa ,Hawaii - Abstract
Coral bleaching, precipitated by the expulsion of the algal symbionts that provide colonies with fixed carbon is a global threat to reef survival. To protect corals from anthropogenic stress, portable tools are needed to detect and diagnose stress syndromes and assess population health prior to extensive bleaching. Here, medical grade Urinalysis strips, used to detect an array of disease markers in humans, were tested on the lab stressed Hawaiian coral species, Montipora capitata (stress resistant) and Pocillopora acuta (stress sensitive), as well as samples from nature that also included Porites compressa. Of the 10 diagnostic reagent tests on these strips, two appear most applicable to corals: ketone and leukocytes. The test strip results from M. capitata were explored using existing transcriptomic data from the same samples and provided evidence of the stress syndromes detected by the strips. We designed a 3D printed smartphone holder and image processing software for field analysis of test strips (TestStripDX) and devised a simple strategy to generate color scores for corals (reflecting extent of bleaching) using a smartphone camera (CoralDX). Our approaches provide field deployable methods, that can be improved in the future (e.g., coral-specific stress test strips) to assess reef health using inexpensive tools and freely available software.
- Published
- 2022
6. Glycerol's contribution to lactate production outside of a glucose intermediate in fasting humans
- Author
-
Ankit Shah, Yujue Wang, Xiaoyang Su, and Fredric E. Wondisford
- Subjects
Blood Glucose ,Glycerol ,Endocrinology ,Glucose ,Liver ,Endocrinology, Diabetes and Metabolism ,Gluconeogenesis ,Humans ,Fasting ,Lactic Acid ,Carbon - Abstract
Glycerol is a well-recognized substrate for new glucose production via gluconeogenesis in the liver. However, its carbon contribution to the glycolytic intermediate lactate is not known in humans.Here we infused stable isotope tracersGlycerol and glucose rates of appearance were 2.21 ± 1.42 μmol/kg/min and 7.81 ± 1.15 μmol/kg/min, respectively. Under steady-state conditions, theGiven that lactate itself is a carbon source for gluconeogenesis and tricycarboxylic cycle intermediates, glycerol's ability to donate carbons to lactate may make it quantitatively more important to intermediary metabolism than currently appreciated.
- Published
- 2022
7. Leukemia inhibitory factor drives glucose metabolic reprogramming to promote breast tumorigenesis
- Author
-
Xuetian Yue, Jianming Wang, Chun-yuan Chang, Juan Liu, Xue Yang, Fan Zhou, Xia Qiu, Vrushank Bhatt, Jessie Yanxiang Guo, Xiaoyang Su, Lanjing Zhang, Zhaohui Feng, and Wenwei Hu
- Subjects
endocrine system ,Cancer Research ,urogenital system ,Carcinogenesis ,Immunology ,Breast Neoplasms ,Cell Biology ,Leukemia Inhibitory Factor ,Cellular and Molecular Neuroscience ,Cell Transformation, Neoplastic ,Glucose ,Cell Line, Tumor ,embryonic structures ,Humans ,Female ,Glycolysis ,Proto-Oncogene Proteins c-akt ,reproductive and urinary physiology ,hormones, hormone substitutes, and hormone antagonists - Abstract
LIF, a multifunctional cytokine, is frequently overexpressed in many types of solid tumors, including breast cancer, and plays an important role in promoting tumorigenesis. Currently, how LIF promotes tumorigenesis is not well-understood. Metabolic reprogramming is a hallmark of cancer cells and a key contributor to cancer progression. However, the role of LIF in cancer metabolic reprogramming is unclear. In this study, we found that LIF increases glucose uptake and drives glycolysis, contributing to breast tumorigenesis. Blocking glucose uptake largely abolishes the promoting effect of LIF on breast tumorigenesis. Mechanistically, LIF overexpression enhances glucose uptake via activating the AKT/GLUT1 axis to promote glycolysis. Blocking the AKT signaling by shRNA or its inhibitors greatly inhibits glycolysis driven by LIF and largely abolishes the promoting effect of LIF on breast tumorigenesis. These results demonstrate an important role of LIF overexpression in glucose metabolism reprogramming in breast cancers, which contributes to breast tumorigenesis. This study also reveals an important mechanism underlying metabolic reprogramming of breast cancers, and identifies LIF and its downstream signaling as potential therapeutic targets for breast cancers, especially those with LIF overexpression.
- Published
- 2021
8. Triterpenoid ursolic acid drives metabolic rewiring and epigenetic reprogramming in treatment/prevention of human prostate cancer
- Author
-
Davit Sargsyan, Hsiao-Chen Dina Kuo, Shanyi Li, Ah-Ng Tony Kong, Xi Zheng, Yujue Wang, Xiaoyang Su, Renyi Wu, and Lujing Wang
- Subjects
Male ,Cancer Research ,Biology ,Article ,Epigenesis, Genetic ,Transcriptome ,Mice ,Metabolomics ,Cell Line, Tumor ,Animals ,Humans ,Epigenetics ,Promoter Regions, Genetic ,Molecular Biology ,Epigenomics ,Sequence Analysis, RNA ,Prostatic Neoplasms ,Epigenome ,DNA Methylation ,Xenograft Model Antitumor Assays ,Triterpenes ,Gene Expression Regulation, Neoplastic ,Oxidative Stress ,Differentially methylated regions ,DNA methylation ,Cancer research ,Reprogramming ,Metabolic Networks and Pathways - Abstract
Ursolic acid (UA) is a triterpenoid phytochemical with a strong anticancer effect. The metabolic rewiring, epigenetic reprogramming, and chemopreventive effect of UA in prostate cancer (PCa) remain unknown. Herein, we investigated the efficacy of UA in PCa xenograft, and its biological effects on cellular metabolism, DNA methylation, and transcriptomic using multi-omics approaches. The metabolomics was quantified by liquid-chromatography-mass spectrometry (LC-MS) while epigenomic CpG methylation in parallel with transcriptomic gene expression was studied by next-generation sequencing technologies. UA administration attenuated the growth of transplanted human VCaP-Luc cells in immunodeficient mice. UA regulated several cellular metabolites and metabolism-related signaling pathways including S-adenosylmethionine (SAM), methionine, glucose 6-phosphate, CDP-choline, phosphatidylcholine biosynthesis, glycolysis, and nucleotide sugars metabolism. RNA-seq analyses revealed UA regulated several signaling pathways, including CXCR4 signaling, cancer metastasis signaling, and NRF2-mediated oxidative stress response. Epigenetic reprogramming study with DNA Methyl-seq uncovered a list of differentially methylated regions (DMRs) associated with UA treatment. Transcriptome-DNA methylome correlative analysis uncovered a list of genes, of which changes in gene expression correlated with the promoter CpG methylation status. Altogether, our results suggest that UA regulates metabolic rewiring of metabolism including SAM potentially driving epigenetic CpG methylation reprogramming, and transcriptomic signaling resulting in the overall anticancer chemopreventive effect.
- Published
- 2021
9. Mitochondrial inhibitors circumvent adaptive resistance to venetoclax and cytarabine combination therapy in acute myeloid leukemia
- Author
-
Jean-Emmanuel Sarry, Thomas Farge, Eléonore Kaphan, Emeline Boet, Francois Vergez, Marie Sabatier, Jérôme Kluza, Nathalie Nicot, Yujue Wang, Andrew H. Wei, Aurélie Bousard, Noémie Gadaud, Pierre-Luc Mouchel, Ambrine Sahal, Nesrine Aroua, Ing Soo Tiong, Nathaniel Polley, Lucille Stuani, Mathilde Gotanègre, Quentin Fovez, Tony Kaoma, Laura Poillet-Perez, Claudie Bosc, Estelle Saland, Marie Tosolini, Guillaume Cognet, Rafael J. Argüello, Florian Rambow, Jean-Jacques Fournié, Christian Recher, Carine Joffre, Clément Larrue, Xiaoyang Su, Jean-Christophe Marine, Jerome Tamburini, Céline Mazzotti, Muriel Picard, Hervé Avet-Loiseau, Florence Cabon, Latifa Jarrou, Centre de Recherches en Cancérologie de Toulouse (CRCT), Université Toulouse III - Paul Sabatier (UT3), Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), LabEx Toucan, LabEx Toucan - Toulouse, Leuven Center for Cancer Biology (VIB-KU-CCB), Catholic University of Leuven - Katholieke Universiteit Leuven (KU Leuven)-Vlaams Instituut voor Biotechnologie [Ghent, Belgique] (VIB), Université Toulouse III Paul Sabatier - Faculté de médecine Purpan (UTPS), Université de Toulouse (UT)-Université de Toulouse (UT), Institut Universitaire du Cancer de Toulouse - Oncopole (IUCT Oncopole - UMR 1037), Université de Toulouse (UT)-Université de Toulouse (UT)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de recherche Cardio-Thoracique de Bordeaux [Bordeaux] (CRCTB), Université Bordeaux Segalen - Bordeaux 2-CHU Bordeaux [Bordeaux]-Institut National de la Santé et de la Recherche Médicale (INSERM), Pôle Anesthésie Réanimation [CHU de Toulouse], Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), Luxembourg Institute of Health (LIH), Rutgers cancer institute of New Jersey [Newark, NJ], Cancer Heterogeneity, Plasticity and Resistance to Therapies - UMR 9020 - U 1277 (CANTHER), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille)-Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université de Genève = University of Geneva (UNIGE), and The Alfred Hospital and Monash University - Department of Clinical Haematology - Melbourne
- Subjects
Cancer Research ,Combination therapy ,[SDV]Life Sciences [q-bio] ,Cell ,Oxidative phosphorylation ,chemistry.chemical_compound ,In vivo ,hemic and lymphatic diseases ,Medicine ,Humans ,ComputingMilieux_MISCELLANEOUS ,Sulfonamides ,business.industry ,Venetoclax ,Cytarabine ,Myeloid leukemia ,Pyruvate dehydrogenase complex ,Bridged Bicyclo Compounds, Heterocyclic ,carbohydrates (lipids) ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,chemistry ,Cancer research ,Azacitidine ,business ,medicine.drug - Abstract
Therapy resistance represents a major clinical challenge in acute myeloid leukemia (AML). Here we define a ‘MitoScore’ signature, which identifies high mitochondrial oxidative phosphorylation in vivo and in patients with AML. Primary AML cells with cytarabine (AraC) resistance and a high MitoScore relied on mitochondrial Bcl2 and were highly sensitive to venetoclax (VEN) + AraC (but not to VEN + azacytidine). Single-cell transcriptomics of VEN + AraC-residual cell populations revealed adaptive resistance associated with changes in oxidative phosphorylation, electron transport chain complex and the TP53 pathway. Accordingly, treatment of VEN + AraC-resistant AML cells with electron transport chain complex inhibitors, pyruvate dehydrogenase inhibitors or mitochondrial ClpP protease agonists substantially delayed relapse following VEN + AraC. These findings highlight the central role of mitochondrial adaptation during AML therapy and provide a scientific rationale for alternating VEN + azacytidine with VEN + AraC in patients with a high MitoScore and to target mitochondrial metabolism to enhance the sensitivity of AML cells to currently approved therapies. Sarry and colleagues demonstrate that adaptive resistance to venetoclax + cytarabine therapy in acute myeloid leukemia relies on mitochondrial respiration and show that combination with electron transport chain complex inhibitors delays relapse in patient-derived xenograft models in vivo.
- Published
- 2021
10. A rare case of right upper lung cancer with azygos lobe and partial anomalous pulmonary venous return
- Author
-
Xiaoyang Su, Qianzhun Huang, Zhiqiang Luo, Ning Fang, and Jian Huang
- Subjects
Pulmonary and Respiratory Medicine ,Lung Neoplasms ,Pulmonary Veins ,Scimitar Syndrome ,Humans ,Surgery ,General Medicine ,Cardiology and Cardiovascular Medicine - Abstract
Background The azygos lobe (AL) combined with partial anomalous pulmonary venous return (PAPVR) is comparatively uncommon as well as in radical surgery for right lung cancer. Case presentation We herein present an extremely rare case of lung cancer coexisting with AL and asymptomatic PAPVR, which was diagnosed with preoperative contrast three-dimensional reconstruction and received radical surgery by thoracoscopy. During the surgery, we preserved azygos vein successfully and found a split type of PAPVR in right upper pulmonary vein. Conclusions AL combined with PAPVR may cause confusion on the vascular separation and disconnection of the right pulmonary hilar. However, preoperative 3D reconstruction is more conducive to the correct performing of this type of surgery.
- Published
- 2021
11. Lactobacillus rhamnosus GG modifies the metabolome of pathobionts in gnotobiotic mice
- Author
-
Lee J. Kerkhof, Ian Nadler, Sheila Bandyopadhyay, Jinhee Kim, Danielle Harlan, Rajbir Singh, Yuling He, Amanda Bumber, Ronaldo P. Ferraris, Nan Gao, Xiaoyang Su, and Iyshwarya Balasubramanian
- Subjects
Male ,Microbiology (medical) ,Staphylococcus aureus ,Metabolite ,Firmicutes ,Gut flora ,medicine.disease_cause ,Microbiology ,law.invention ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,Probiotic ,Propionibacterium acnes ,chemistry.chemical_compound ,Fecal metabolites ,Lactobacillus rhamnosus ,law ,Competitive exclusion ,Metabolome ,medicine ,Animals ,Germ-Free Life ,Humans ,Liquid chromatography, mass spectrometry ,Gram-Positive Bacterial Infections ,030304 developmental biology ,Inflammation ,0303 health sciences ,biology ,Lacticaseibacillus rhamnosus ,030306 microbiology ,Probiotics ,Microbiota ,biology.organism_classification ,QR1-502 ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,chemistry ,Cytokines ,Female ,Germ-free mice ,Research Article - Abstract
Background Lactobacillus rhamnosus GG (LGG) is the most widely used probiotic, but the mechanisms underlying its beneficial effects remain unresolved. Previous studies typically inoculated LGG in hosts with established gut microbiota, limiting the understanding of specific impacts of LGG on host due to numerous interactions among LGG, commensal microbes, and the host. There has been a scarcity of studies that used gnotobiotic animals to elucidate LGG-host interaction, in particular for gaining specific insights about how it modifies the metabolome. To evaluate whether LGG affects the metabolite output of pathobionts, we inoculated with LGG gnotobiotic mice containing Propionibacterium acnes, Turicibacter sanguinis, and Staphylococcus aureus (PTS). Results 16S rRNA sequencing of fecal samples by Ion Torrent and MinION platforms showed colonization of germ-free mice by PTS or by PTS plus LGG (LTS). Although the body weights and feeding rates of mice remained similar between PTS and LTS groups, co-associating LGG with PTS led to a pronounced reduction in abundance of P. acnes in the gut. Addition of LGG or its secretome inhibited P. acnes growth in culture. After optimizing procedures for fecal metabolite extraction and metabolomic liquid chromatography-mass spectrometry analysis, unsupervised and supervised multivariate analyses revealed a distinct separation among fecal metabolites of PTS, LTS, and germ-free groups. Variables-important-in-projection scores showed that LGG colonization robustly diminished guanine, ornitihine, and sorbitol while significantly elevating acetylated amino acids, ribitol, indolelactic acid, and histamine. In addition, carnitine, betaine, and glutamate increased while thymidine, quinic acid and biotin were reduced in both PTS and LTS groups. Furthermore, LGG association reduced intestinal mucosal expression levels of inflammatory cytokines, such as IL-1α, IL-1β and TNF-α. Conclusions LGG co-association had a negative impact on colonization of P. acnes, and markedly altered the metabolic output and inflammatory response elicited by pathobionts.
- Published
- 2021
12. Autophagy modulates lipid metabolism to maintain metabolic flexibility for Lkb1-deficient Kras-driven lung tumorigenesis
- Author
-
Jessie Yanxiang Guo, Wali Kamran, Zhixian Sherrie Hu, Amy Lee, Khoosheh Khayati, Vrushank Bhatt, and Xiaoyang Su
- Subjects
Lung Neoplasms ,Carcinogenesis ,Cell Survival ,Tumor initiation ,Biology ,medicine.disease_cause ,Autophagy-Related Protein 7 ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Autophagy ,Genetics ,medicine ,Animals ,Humans ,Beta oxidation ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,0303 health sciences ,Kinase ,Intracellular Signaling Peptides and Proteins ,Lipid metabolism ,Lipid Metabolism ,Cell biology ,Disease Models, Animal ,030220 oncology & carcinogenesis ,Cancer cell ,Carrier Proteins ,Energy Metabolism ,Gene Deletion ,Homeostasis ,Research Paper ,Developmental Biology - Abstract
Loss of tumor suppressor liver kinase B1 (LKB1) promotes cancer cell proliferation but also leads to decreased metabolic plasticity in dealing with energy crises. Autophagy is a protective process involving self-cannibalization to maintain cellular energy homeostasis during nutrient deprivation. We developed a mouse model for Lkb1-deficient lung cancer with conditional deletion of essential autophagy gene Atg7 to test whether autophagy compensates for LKB1 loss for tumor cells to survive energy crises. We found that autophagy ablation was synthetically lethal during Lkb1-deficient lung tumorigenesis in both tumor initiation and tumor growth. We further found that autophagy deficiency causes defective intracellular recycling, which limits amino acids to support mitochondrial energy production in starved cancer cells and causes autophagy-deficient cells to be more dependent on fatty acid oxidation (FAO) for energy production, leading to reduced lipid reserve and energy crisis. Our findings strongly suggest that autophagy inhibition could be a strategy for treating LKB1-deficient lung tumors.
- Published
- 2019
13. Glycerate from intestinal fructose metabolism induces islet cell damage and glucose intolerance
- Author
-
Yanru Wu, Chi Wut Wong, Eric N. Chiles, Allyson L. Mellinger, Hosung Bae, Sunhee Jung, Ted Peterson, Jamie Wang, Marcos Negrete, Qiang Huang, Lihua Wang, Cholsoon Jang, David C. Muddiman, Xiaoyang Su, Ian Williamson, and Xiling Shen
- Subjects
Blood Glucose ,Physiology ,Cell Biology ,Fructose ,Diet, High-Fat ,Dietary Fats ,Article ,Islets of Langerhans ,Glucose ,Diabetes Mellitus, Type 2 ,Glucose Intolerance ,Humans ,Insulin ,Molecular Biology - Abstract
Dietary fructose, especially in the context of a high-fat western diet, has been linked to type 2 diabetes. Although the effect of fructose on liver metabolism has been extensively studied, a significant portion of the fructose is first metabolized in the small intestine. Here, we report that dietary fat enhances intestinal fructose metabolism, which releases glycerate into the blood. Chronic high systemic glycerate levels induce glucose intolerance by slowly damaging pancreatic islet cells and reducing islet sizes. Our findings provide a link between dietary fructose and diabetes that is modulated by dietary fat.
- Published
- 2021
14. The nuclear receptor HNF4 drives a brush border gene program conserved across murine intestine, kidney, and embryonic yolk sac
- Author
-
Weihuan Cao, Joseph Hur, Christopher E. Ellison, Raj Malhotra, Roshan P. Vasoya, Abigail Dupre, Min Yang, Juan Flores, Michael P. Verzi, Amrik Sahota, Aditya Parthasarathy, Lei Chen, Eric Chiles, Rohit Aita, Nan Gao, Natalie H. Toke, Shirley Luo, Edward M. Bonder, and Xiaoyang Su
- Subjects
0301 basic medicine ,Cell biology ,Brush border ,Science ,Receptors, Cytoplasmic and Nuclear ,General Physics and Astronomy ,Mice, Transgenic ,Biology ,Kidney ,Article ,Epithelium ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Microscopy, Electron, Transmission ,Animals ,Humans ,Intestinal Mucosa ,Enhancer ,Transcription factor ,Yolk Sac ,Mice, Knockout ,Regulation of gene expression ,Multidisciplinary ,Microvilli ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Kidney metabolism ,Promoter ,General Chemistry ,Gene regulation ,Chromatin ,body regions ,Intestines ,030104 developmental biology ,Gene Expression Regulation ,Hepatocyte Nuclear Factor 4 ,Hepatocyte nuclear factor 4 ,embryonic structures ,030217 neurology & neurosurgery - Abstract
The brush border is comprised of microvilli surface protrusions on the apical surface of epithelia. This specialized structure greatly increases absorptive surface area and plays crucial roles in human health. However, transcriptional regulatory networks controlling brush border genes are not fully understood. Here, we identify that hepatocyte nuclear factor 4 (HNF4) transcription factor is a conserved and important regulator of brush border gene program in multiple organs, such as intestine, kidney and yolk sac. Compromised brush border gene signatures and impaired transport were observed in these tissues upon HNF4 loss. By ChIP-seq, we find HNF4 binds and activates brush border genes in the intestine and kidney. H3K4me3 HiChIP-seq identifies that HNF4 loss results in impaired chromatin looping between enhancers and promoters at gene loci of brush border genes, and instead enhanced chromatin looping at gene loci of stress fiber genes in the intestine. This study provides comprehensive transcriptional regulatory mechanisms and a functional demonstration of a critical role for HNF4 in brush border gene regulation across multiple murine epithelial tissues., Brush border gene regulation in various different tissues is incompletely understood. Here, the authors show HNF4 regulates the brush border gene program in multiple organs, such as intestine, kidney and yolk sac, and also intestinal chromatin looping in these tissues between promoters and enhancers.
- Published
- 2021
15. Parkin ubiquitinates phosphoglycerate dehydrogenase to suppress serine synthesis and tumor progression
- Author
-
Xuetian Yue, Wenwei Hu, Zhaohui Feng, Cen Zhang, Bruce G. Haffty, Juan Liu, Xiao Xin Sun, Xiaoyang Su, Yanchen Li, Shan Huang, Shou-En Lu, Hao Wu, Eileen White, and Zhiyuan Shen
- Subjects
0301 basic medicine ,Ubiquitin-Protein Ligases ,medicine.disease_cause ,Parkin ,Serine ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Phosphoglycerate dehydrogenase ,Cell Proliferation ,biology ,Chemistry ,Parkinson Disease ,General Medicine ,Ubiquitin ligase ,nervous system diseases ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research ,Carcinogenesis ,Research Article - Abstract
Phosphoglycerate dehydrogenase (PHGDH), the first rate-limiting enzyme of serine synthesis, is frequently overexpressed in human cancer. PHGDH overexpression activates serine synthesis to promote cancer progression. Currently, PHGDH regulation in normal cells and cancer is not well understood. Parkin, an E3 ubiquitin ligase involved in Parkinson's disease, is a tumor suppressor. Parkin expression is frequently downregulated in many types of cancer, and its tumor-suppressive mechanism is poorly defined. Here, we show that PHGDH is a substrate for Parkin-mediated ubiquitination and degradation. Parkin interacted with PHGDH and ubiquitinated PHGDH at lysine 330, leading to PHGDH degradation to suppress serine synthesis. Parkin deficiency in cancer cells stabilized PHGDH and activated serine synthesis to promote cell proliferation and tumorigenesis, which was largely abolished by targeting PHGDH with RNA interference, CRISPR/Cas9 KO, or small-molecule PHGDH inhibitors. Furthermore, Parkin expression was inversely correlated with PHGDH expression in human breast cancer and lung cancer. Our results revealed PHGDH ubiquitination by Parkin as a crucial mechanism for PHGDH regulation that contributes to the tumor-suppressive function of Parkin and identified Parkin downregulation as a critical mechanism underlying PHGDH overexpression in cancer.
- Published
- 2020
16. Transcriptome-wide Association Study Identifies Genetically Dysregulated Genes in Diabetic Neuropathy
- Author
-
Sicheng Du, Danfeng Lan, Xiaoyang Su, Deng-Feng Zhang, Ying Li, Qiuping Yang, Rui Bi, Yan Zhao, and Hong-Yan Jiang
- Subjects
Diabetic neuropathy ,Genome-wide association study ,Bioinformatics ,Transcriptome ,Mice ,Diabetic Neuropathies ,Diabetes mellitus ,Drug Discovery ,medicine ,Animals ,Humans ,Tibial nerve ,business.industry ,Organic Chemistry ,General Medicine ,MERTK ,medicine.disease ,Computer Science Applications ,Disease Models, Animal ,Diabetes Mellitus, Type 2 ,Expression quantitative trait loci ,Sciatic nerve ,business ,Algorithms ,Genome-Wide Association Study - Abstract
Background: Complications are the main cause of the disease burden of diabetes. Genes determining the development and progression of diabetic complications remain to be identified. Diabetic neuropathy is the most common and debilitating complication and mainly affects the nerves of legs and feet. In this study, we attempted to identify diabetic neuropathy-specific genes from reliable large-scale genome-wide association studies (GWASs) for diabetes perse. Methods: Taking advantage of publicly available data, we initially converted the GWAS signals to transcriptomic profiles in the tibial nerve using the functional summary-based imputation (FUSION) algorithm. The FUSION-derived genes were then checked to determine whether they were differentially expressed in the sciatic nerve of mouse models of diabetic neuropathy. The dysregulated genes identified in the sciatic nerve were explored in the blood of patients with diabetes. Results: We found that eleven out of 452 FUSION-derived genes were regulated by diabetes GWAS loci and were altered in the sciatic nerve of mouse models with early-stage neuropathy. Among the eleven genes, significant (P-value#60;0.05) expression alterations of HSD17B4, DHX32, MERTK, and SFXN4 could be detected in the blood of human patients. Conclusions: Our analyses identified genes with an effect in the sciatic nerve and provided the possibility of noninvasive early detection of diabetic neuropathy.
- Published
- 2020
17. G protein-coupled kisspeptin receptor induces metabolic reprograming and tumorigenesis in estrogen receptor-negative breast cancer
- Author
-
Mai Uyen Nguyen, Ali Abbara, Muriel Brackstone, Paul Bech, Wei-Xing Zong, Andy V. Babwah, Sharon R. Pine, Sophie A Clarke, Stephania Guzman, Frederic E. Wondisford, Waljit S. Dhillo, David A. Hess, Cameron Goertzen, Xiaoyang Su, Magdalena Dragan, Alan B. Tuck, and Moshmi Bhattacharya
- Subjects
Cancer Research ,Carcinogenesis ,Glutamine ,Estrogen receptor ,Triple Negative Breast Neoplasms ,Mice, SCID ,0601 Biochemistry and Cell Biology ,medicine.disease_cause ,Metastasis ,Malignant transformation ,Breast cancer ,0302 clinical medicine ,Kisspeptin ,Cell Movement ,Mice, Inbred NOD ,0303 health sciences ,lcsh:Cytology ,Nucleotides ,Middle Aged ,Cellular Reprogramming ,Cancer metabolism ,Tumor Burden ,3. Good health ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Female ,Signal Transduction ,Adult ,Immunology ,Biology ,Article ,Proto-Oncogene Proteins c-myc ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Glutaminase ,Cell Line, Tumor ,Progesterone receptor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,1112 Oncology and Carcinogenesis ,lcsh:QH573-671 ,Aged ,Cell Proliferation ,030304 developmental biology ,Glutaminolysis ,Cell Biology ,medicine.disease ,Case-Control Studies ,Cancer research ,Energy Metabolism ,Receptors, Kisspeptin-1 - Abstract
Triple-negative breast cancer (TNBC) is a highly metastatic and deadly disease. TNBC tumors lack estrogen receptor (ERα), progesterone receptor (PR), and HER2 (ErbB2) and exhibit increased glutamine metabolism, a requirement for tumor growth. The G protein-coupled kisspeptin receptor (KISS1R) is highly expressed in patient TNBC tumors and promotes malignant transformation of breast epithelial cells. This study found that TNBC patients displayed elevated plasma kisspeptin levels compared with healthy subjects. It also provides the first evidence that in addition to promoting tumor growth and metastasis in vivo, KISS1R-induced glutamine dependence of tumors. In addition, tracer-based metabolomics analyses revealed that KISS1R promoted glutaminolysis and nucleotide biosynthesis by increasing c-Myc and glutaminase levels, key regulators of glutamine metabolism. Overall, this study establishes KISS1R as a novel regulator of TNBC metabolism and metastasis, suggesting that targeting KISS1R could have therapeutic potential in the treatment of TNBC.
- Published
- 2020
18. In-Source CID Ramping (InCIDR) and Co-Variant Ion Analysis of Hydrophilic Interaction Chromatography (HILIC) Metabolomics
- Author
-
Eric Chiles, Xiaoyang Su, Sara Maimouni, Wei-Xing Zong, Chi Song, and Fredric E. Wondisford
- Subjects
Chromatography ,Chemistry ,Hydrophilic interaction chromatography ,Article ,Mass Spectrometry ,Metabolomics data ,Adduct ,Metabolomics ,Cell Line, Tumor ,Redundancy (engineering) ,Humans ,Hydrophobic and Hydrophilic Interactions ,Chromatography, High Pressure Liquid - Abstract
A large proportion of the complexity and redundancy of LC-MS metabolomics data comes from adduct formation. To reduce such redundancy, many tools have been developed to recognize and annotate adduct ions. These tools rely on pre-defined adduct lists which are learned empirically from reverse phase LC-MS studies. Meanwhile, hydrophilic interaction chromatography (HILIC) is gaining popularity in metabolomics studies due to better performance on polar compounds. HILIC methods typically use high concentration of buffer salts for improved chromatography performance. It is therefore necessary to analyze the adduct formation in HILIC metabolomics. To this end, we developed co-variant ion analysis (COVINA) to investigate the metabolite adduct formation. Using this tool, we completely annotated 201 adduct and fragment ions of 10 metabolites. Many of the metabolite adduct ions are found to contain cluster ions of mobile phase additives. We further utilized COVINA to find the major ionization forms of metabolites. Our results show that for some metabolites the adduct ion signals can be >200-fold higher than the deprotonated form, offering better sensitivity for targeted metabolomics analysis. Finally, we developed the in-source CID ramping (InCIDR) method to analyze the intensity changes of the adduct and fragment ions of the metabolites. Our analysis demonstrates a promising method to distinguish the protonated/deprotonated ions of the metabolites from the adduct and fragment ions.
- Published
- 2020
19. Serine Catabolism Feeds NADH when Respiration Is Impaired
- Author
-
Cholsoon Jang, Raphael J. Morscher, Lin Wang, Zhaoyue Zhang, Joshua D. Rabinowitz, Lifeng Yang, Shilpy Joshi, Lingfan Liang, Zhixian Hu, Melanie R. McReynolds, Zihong Chen, Jonathan M. Ghergurovich, Le Zhan, Eileen White, Juan Carlos Garcia Canaveras, Xiaoyang Su, and Johannes A. Mayr
- Subjects
0301 basic medicine ,Physiology ,Nude ,complex I inhibitor ,Mice, Nude ,Medical Biochemistry and Metabolomics ,serine hydroxymethyltransferase ,Inbred C57BL ,Cell Line ,respiration inhibition ,Serine ,Mice ,Endocrinology & Metabolism ,03 medical and health sciences ,0302 clinical medicine ,Respiration ,Animals ,Humans ,Molecular Biology ,methylene tetrahydrofolate dehydrogenase ,MTHFD2 ,Chemistry ,Catabolism ,Cell growth ,hypoxia ,Cell Biology ,NAD ,SHMT2 ,Electron transport chain ,Cell Hypoxia ,Mitochondria ,Mice, Inbred C57BL ,Oxygen ,Glutamine ,mitochondrial disease ,030104 developmental biology ,serine catabolism ,Biochemistry ,Serine hydroxymethyltransferase ,redox ,NADH ,Biochemistry and Cell Biology ,NAD+ kinase ,030217 neurology & neurosurgery - Abstract
NADH provides electrons for aerobic ATP production. In cells deprived of oxygen or with impaired electron transport chain activity, NADH accumulation can be toxic. To minimize such toxicity, elevated NADH inhibits the classical NADH producing pathways: glucose, glutamine, and fat oxidation. Here, through deuterium tracing studies in cultured cells and mice, we show that folate-dependent serine catabolism also produces substantial NADH. Strikingly, when respiration is impaired, serine catabolism through methylene tetrahydrofolate dehydrogenase (MTHFD2) becomes a major NADH source. In cells whose respiration is slowed by hypoxia, metformin, or genetic lesions, mitochondrial serine catabolism inhibition partially normalizes NADH levels and facilitates cell growth. In mice with engineered mitochondrial complex I deficiency (NDUSF4-/-), serine’s contribution to NADH is elevated and progression of spasticity is modestly slowed by pharmacological blockade of serine degradation. Thus, when respiration is impaired, serine catabolism contributes to toxic NADH accumulation.
- Published
- 2020
- Full Text
- View/download PDF
20. Autophagy maintains tumor growth through circulating arginine
- Author
-
Janice M. Mehnert, Haiyan Zheng, Cherry Jiang, Le Zhan, Anurag Maganti, Yang Yang, Jessie Yanxiang Guo, Laura Poillet-Perez, Daniel W. Sharp, Edmund C. Lattime, Xiaoqi Xie, Xiaoyang Su, Zhixian Sherrie Hu, Eileen White, Marcus Bosenberg, Joshua D. Rabinowitz, and Wenyun Lu
- Subjects
0301 basic medicine ,Male ,Ornithine ,Arginine ,Carcinogenesis ,ATG5 ,Ornithine transcarbamylase ,Biology ,Autophagy-Related Protein 7 ,Article ,Autophagy-Related Protein 5 ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Neoplasms ,Autophagy ,Animals ,Humans ,ARG1 ,Cell Proliferation ,2. Zero hunger ,Multidisciplinary ,Arginase ,Allografts ,3. Good health ,Diet ,030104 developmental biology ,Editorial ,chemistry ,Liver ,Dietary Supplements ,Cancer research ,Hepatocytes ,Neoplasm Transplantation - Abstract
Autophagy captures intracellular components and delivers them to lysosomes, where they are degraded and recycled to sustain metabolism and to enable survival during starvation1-5. Acute, whole-body deletion of the essential autophagy gene Atg7 in adult mice causes a systemic metabolic defect that manifests as starvation intolerance and gradual loss of white adipose tissue, liver glycogen and muscle mass1. Cancer cells also benefit from autophagy. Deletion of essential autophagy genes impairs the metabolism, proliferation, survival and malignancy of spontaneous tumours in models of autochthonous cancer6,7. Acute, systemic deletion of Atg7 or acute, systemic expression of a dominant-negative ATG4b in mice induces greater regression of KRAS-driven cancers than does tumour-specific autophagy deletion, which suggests that host autophagy promotes tumour growth1,8. Here we show that host-specific deletion of Atg7 impairs the growth of multiple allografted tumours, although not all tumour lines were sensitive to host autophagy status. Loss of autophagy in the host was associated with a reduction in circulating arginine, and the sensitive tumour cell lines were arginine auxotrophs owing to the lack of expression of the enzyme argininosuccinate synthase 1. Serum proteomic analysis identified the arginine-degrading enzyme arginase I (ARG1) in the circulation of Atg7-deficient hosts, and in vivo arginine metabolic tracing demonstrated that serum arginine was degraded to ornithine. ARG1 is predominantly expressed in the liver and can be released from hepatocytes into the circulation. Liver-specific deletion of Atg7 produced circulating ARG1, and reduced both serum arginine and tumour growth. Deletion of Atg5 in the host similarly regulated [corrected] circulating arginine and suppressed tumorigenesis, which demonstrates that this phenotype is specific to autophagy function rather than to deletion of Atg7. Dietary supplementation of Atg7-deficient hosts with arginine partially restored levels of circulating arginine and tumour growth. Thus, defective autophagy in the host leads to the release of ARG1 from the liver and the degradation of circulating arginine, which is essential for tumour growth; this identifies a metabolic vulnerability of cancer.
- Published
- 2018
21. mTORC2 modulates the amplitude and duration of GFAT1 Ser-243 phosphorylation to maintain flux through the hexosamine pathway during starvation
- Author
-
Sharon Liu, Peter K. Kim, Austin Chang, Chadni Patel, Danielle Albaciete, Xiaoyang Su, Swati Rajput, Estela Jacinto, Joseph G. Moloughney, Guy Werlen, Cedric Magaway, Nicole M. Vega-Cotto, and Chang Chih Wu
- Subjects
0301 basic medicine ,Mechanistic Target of Rapamycin Complex 2 ,Biochemistry ,mTORC2 ,Acetylglucosamine ,03 medical and health sciences ,Serine ,Animals ,Humans ,Protein phosphorylation ,Phosphorylation ,Molecular Biology ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) ,Uridine Diphosphate N-Acetylglucosamine ,biology ,Chemistry ,AMPK ,Hexosamines ,Cell Biology ,Biosynthetic Pathways ,Cell biology ,Glutamine ,Metabolic pathway ,030104 developmental biology ,Starvation ,biology.protein ,Signal Transduction - Abstract
The mechanistic target of rapamycin (mTOR) controls metabolic pathways in response to nutrients. Recently, we have shown that mTOR complex 2 (mTORC2) modulates the hexosamine biosynthetic pathway (HBP) by promoting the expression of the key enzyme of the HBP, glutamine:fructose-6-phosphate aminotransferase 1 (GFAT1). Here, we found that GFAT1 Ser-243 phosphorylation is also modulated in an mTORC2-dependent manner. In response to glutamine limitation, active mTORC2 prolongs the duration of Ser-243 phosphorylation, albeit at lower amplitude. Blocking glycolysis using 2-deoxyglucose robustly enhances Ser-243 phosphorylation, correlating with heightened mTORC2 activation, increased AMPK activity, and O-GlcNAcylation. However, when 2-deoxyglucose is combined with glutamine deprivation, GFAT1 Ser-243 phosphorylation and mTORC2 activation remain elevated, whereas AMPK activation and O-GlcNAcylation diminish. Phosphorylation at Ser-243 promotes GFAT1 expression and production of GFAT1-generated metabolites including ample production of the HBP end-product, UDP-GlcNAc, despite nutrient starvation. Hence, we propose that the mTORC2-mediated increase in GFAT1 Ser-243 phosphorylation promotes flux through the HBP to maintain production of UDP-GlcNAc when nutrients are limiting. Our findings provide insights on how the HBP is reprogrammed via mTORC2 in nutrient-addicted cancer cells.
- Published
- 2018
22. Local production of lactate, ribose phosphate, and amino acids by human triple-negative breast cancer
- Author
-
Aaron Killian, Maren K Levin, Natalia Briones, Alison Barron, Jeffrey P. Lamont, Matthew J. McBride, William P.D. Hendricks, Tuoc Dao, Jonathan M. Ghergurovich, Virginia Espina, Claudius Mueller, Xiaoyang Su, Salvatore Facista, Joyce O'Shaughnessy, Alexis J. Cowan, Jessica D. Lang, Joshua D. Rabinowitz, Esther San Roman Rodriguez, and Daniel D. Von Hoff
- Subjects
Proteomics ,chemistry.chemical_classification ,Lung Neoplasms ,Anabolism ,Catabolism ,Triple Negative Breast Neoplasms ,General Medicine ,Metabolism ,Pentose phosphate pathway ,Article ,Amino acid ,Glutamine ,Glucose ,chemistry ,Biochemistry ,Carcinoma, Non-Small-Cell Lung ,Humans ,Glycolysis ,Lactic Acid ,Ribosemonophosphates ,Amino Acids ,Amino acid synthesis - Abstract
Summary Background Upregulated glucose metabolism is a common feature of tumors. Glucose can be broken down by either glycolysis or the oxidative pentose phosphate pathway (oxPPP). The relative usage within tumors of these catabolic pathways remains unclear. Similarly, the extent to which tumors make biomass precursors from glucose, versus take them up from the circulation, is incompletely defined. Methods We explore human triple-negative breast cancer (TNBC) metabolism by isotope tracing with [1,2-13C]glucose, a tracer that differentiates glycolytic versus oxPPP catabolism and reveals glucose-driven anabolism. Patients enrolled in clinical trial NCT03457779 and received intravenous (i.v.) infusion of [1,2-13C]glucose during core biopsy of their primary TNBC. Tumor samples were analyzed for metabolite labeling by liquid chromatography-mass spectrometry (LC-MS). Genomic and proteomic analyses were performed and related to observed metabolic fluxes. Findings TNBC ferments glucose to lactate, with glycolysis dominant over the oxPPP. Most ribose phosphate is nevertheless produced by oxPPP. Glucose also feeds amino acid synthesis, including of serine, glycine, aspartate, glutamate, proline, and glutamine (but not asparagine). Downstream in glycolysis, tumor pyruvate and lactate labeling exceed that found in serum, indicating that lactate exchange via monocarboxylic transporters is less prevalent in human TNBC compared with most normal tissues or non-small cell lung cancer. Conclusions Glucose directly feeds ribose phosphate, amino acid synthesis, lactate, and the tricarboxylic acid (TCA) cycle locally within human breast tumors. Funding The clinical trial, genomics, and proteomics were funded by the Baylor Scott & White Dallas Foundation, Dallas, Texas. Metabolic analyses were supported by NIH grants 1DP1DK113643, R01CA163591, and P30CA072720.
- Published
- 2021
23. Bisphosphoglycerate mutase controls serine pathway flux via 3-phosphoglycerate
- Author
-
Mark Esposito, Xiaoyang Su, David H. Perlman, Joshua D. Rabinowitz, Yael David, Michael Haugbro, Rob C. Oslund, Jung-Min Kee, Yibin Kang, Eva J. Ge, Tom W. Muir, and Boyuan Wang
- Subjects
0301 basic medicine ,Glyceric Acids ,Article ,Serine ,Phosphoglycerate mutase ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Biosynthesis ,Phosphoglycerate Mutase 1 ,Tumor Cells, Cultured ,Humans ,Phosphoglycerate dehydrogenase ,Molecular Biology ,Bisphosphoglycerate mutase ,Phosphoglycerate Mutase ,Phosphoglycerate kinase ,biology ,Cell Biology ,Phosphoglycerate Mutase Deficiency ,030104 developmental biology ,Biochemistry ,chemistry ,030220 oncology & carcinogenesis ,biology.protein - Abstract
Lower glycolysis involves a series of reversible reactions, which interconvert intermediates that also feed anabolic pathways. 3-phosphoglycerate (3-PG) is an abundant lower glycolytic intermediate that feeds serine biosynthesis via the enzyme phosphoglycerate dehydrogenase, which is genomically amplified in several cancers. Phosphoglycerate mutase (PGAM1) catalyzes the isomerization of 3-PG into the downstream glycolytic intermediate 2-phosphoglycerate (2-PG). Catalytic activity of PGAM1 requires its histidine phosphorylation. We show that the primary PGAM1 histidine phosphate donor is 2,3-bisphosphoglycerate (2,3-BPG), which is made from the glycolytic intermediate 1,3-bisphosphoglycerate (1,3-BPG) by bisphosphoglycerate mutase (BPGM). When BPGM is knocked out, 1,3-BPG can directly phosphorylate PGAM1. In this case, PGAM1 phosphorylation and activity are decreased, but nevertheless sufficient to maintain normal glycolytic flux and cellular growth rate. 3-PG, however, accumulates, leading to increased serine synthesis. Thus, one biological function of BPGM is to control glycolytic intermediate levels and thereby serine biosynthetic flux., Graphical Abstract
- Published
- 2017
24. Quantitative analysis of NAD synthesis-breakdown fluxes
- Author
-
David W. Frederick, Joseph A. Baur, Sheng Hui, Karthikeyani Chellappa, Joshua D. Rabinowitz, William J. Quinn, Timothy J. Mitchison, Le Zhan, Kristin A. Krukenberg, Xiaoyang Su, Ling Liu, Philip Redpath, Marie E. Migaud, and Eileen White
- Subjects
0301 basic medicine ,Niacinamide ,Physiology ,Nicotinamide adenine dinucleotide ,Cofactor ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Intestine, Small ,Animals ,Humans ,Sirtuins ,Muscle, Skeletal ,Molecular Biology ,chemistry.chemical_classification ,biology ,Nicotinamide ,Chemistry ,Tryptophan ,Cell Biology ,Metabolism ,Hep G2 Cells ,HCT116 Cells ,NAD ,Mice, Inbred C57BL ,030104 developmental biology ,Enzyme ,Biochemistry ,Liver ,Nicotinamide riboside ,biology.protein ,Female ,NAD+ kinase ,Poly(ADP-ribose) Polymerases ,Niacin ,Spleen - Abstract
The redox cofactor nicotinamide adenine dinucleotide (NAD) plays a central role in metabolism and is a substrate for signaling enzymes including poly-ADP-ribose-polymerases (PARPs) and sirtuins. NAD concentration falls during aging, which has triggered intense interest in strategies to boost NAD levels. A limitation in understanding NAD metabolism has been reliance on concentration measurements. Here, we present isotope-tracer methods for NAD flux quantitation. In cell lines, NAD was made from nicotinamide and consumed largely by PARPs and sirtuins. In vivo, NAD was made from tryptophan selectively in the liver, which then excreted nicotinamide. NAD fluxes varied widely across tissues, with high flux in the small intestine and spleen and low flux in the skeletal muscle. Intravenous administration of nicotinamide riboside or mononucleotide delivered intact molecules to multiple tissues, but the same agents given orally were metabolized to nicotinamide in the liver. Thus, flux analysis can reveal tissue-specific NAD metabolism.
- Published
- 2018
25. Chemical basis for deuterium labeling of fat and NADPH
- Author
-
Ling Liu, Joshua D. Rabinowitz, Li Chen, Zhaoyue Zhang, and Xiaoyang Su
- Subjects
0301 basic medicine ,Hydrogen ,chemistry.chemical_element ,Flavin group ,Photochemistry ,Biochemistry ,Chemical reaction ,Redox ,Catalysis ,Article ,03 medical and health sciences ,0302 clinical medicine ,Colloid and Surface Chemistry ,Kinetic isotope effect ,Humans ,Chemistry ,Fatty Acids ,Deuterium Exchange Measurement ,Water ,General Chemistry ,Metabolism ,Deuterium ,HCT116 Cells ,030104 developmental biology ,HEK293 Cells ,Adipose Tissue ,Oxidation-Reduction ,030217 neurology & neurosurgery ,NADP - Abstract
Much understanding of metabolism is based on monitoring chemical reactions in cells with isotope tracers. For this purpose, 13C is well suited due to its stable incorporation into bio-molecules and minimal kinetic isotope effect. For redox reactions, however, deuterium tracing can provide valuable additional information. To date, studies examining NADPH production with deuterated carbon sources have failed to account for roughly half of NADPH’s redox active hydrogen. Here we show that the missing hydrogen is the result of enzyme-catalyzed H-D exchange between water and NADPH. While isolated NADPH does not undergo H-D exchange with water, such exchange is catalyzed by Flavin enzymes and occurs rapidly in cells. Correction for H-D exchange is required for accurate assessment of the biological sources of NADPH’s high energy electrons. Deuterated water (D2O) is frequently used to monitor fat synthesis in vivo, but the chemical pathway of the deuteron into fat remains unclear. We show that D2O labels fatty acids primarily via NADPH. Knowledge of this labeling route enables calculation, without any fitting parameters, of the mass isotope distributions of fatty acids from cells grown in D2O. Thus, knowledge of enzyme-catalyzed H-D exchange between water and NADPH enables chemically accurate interpretation of deuterium tracing studies of redox cofactor and fatty acid metabolism.
- Published
- 2017
26. Metabolite Measurement: Pitfalls to Avoid and Practices to Follow
- Author
-
Ian A. Lewis, Oliver Fiehn, Xiaoyang Su, Joshua D. Rabinowitz, Wenyun Lu, and Matthias S. Klein
- Subjects
0301 basic medicine ,Systematic error ,Biochemistry & Molecular Biology ,Magnetic Resonance Spectroscopy ,Liquid Phase Microextraction ,Metabolite ,Structural diversity ,Bioengineering ,Guidelines as Topic ,Medical and Health Sciences ,Biochemistry ,Gas Chromatography-Mass Spectrometry ,Mass Spectrometry ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Rare Diseases ,Metabolomics ,Adenosine Triphosphate ,Affordable and Clean Energy ,Animals ,Humans ,metabonomics ,metabolite extraction ,chemistry.chemical_classification ,Chromatography ,Liquid ,accuracy ,Biomolecule ,Primary metabolite ,stability ,Biological Sciences ,Glutathione ,030104 developmental biology ,chemistry ,mass spectroscopy ,Solvents ,Biochemical engineering ,NADP ,Chromatography, Liquid - Abstract
Metabolites are the small biological molecules involved in energy conversion and biosynthesis. Studying metabolism is inherently challenging due to metabolites’ reactivity, structural diversity, and broad concentration range. Herein, we review the common pitfalls encountered in metabolomics and provide concrete guidelines for obtaining accurate metabolite measurements, focusing on water-soluble primary metabolites. We show how seemingly straightforward sample preparation methods can introduce systematic errors (e.g., owing to interconversion among metabolites) and how proper selection of quenching solvent (e.g., acidic acetonitrile:methanol:water) can mitigate such problems. We discuss the specific strengths, pitfalls, and best practices for each common analytical platform: liquid chromatography-mass spectrometry (LC-MS), gas chromatography-mass spectrometry (GC-MS), nuclear magnetic resonance (NMR), and enzyme assays. Together this information provides a pragmatic knowledge base for carrying out biologically informative metabolite measurements.
- Published
- 2017
27. Dph7 Catalyzes a Previously Unknown Demethylation Step in Diphthamide Biosynthesis
- Author
-
Hening Lin, Xiaoyang Su, Wei Chen, Zhewang Lin, Bo Ci, and Sheng Zhang
- Subjects
Saccharomyces cerevisiae Proteins ,Saccharomyces cerevisiae ,Vesicular Transport Proteins ,Biochemistry ,Methylation ,Catalysis ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,Biosynthesis ,Humans ,Histidine ,Demethylation ,chemistry.chemical_classification ,biology ,Communication ,Diphthamide ,General Chemistry ,biology.organism_classification ,Yeast ,Biosynthetic Pathways ,Enzyme ,chemistry ,Protein Processing, Post-Translational ,Gene Deletion - Abstract
Present on archaeal and eukaryotic translation elongation factor 2, diphthamide represents one of the most intriguing post-translational modifications on proteins. The biosynthesis of diphthamide was proposed to occur in three steps requiring seven proteins, Dph1-7, in eukaryotes. The functional assignments of Dph1-5 in the first and second step have been well established. Recent studies suggest that Dph6 (yeast YLR143W or human ATPBD4) and Dph7 (yeast YBR246W or human WDR85) are involved in the last amidation step, with Dph6 being the actual diphthamide synthetase catalyzing the ATP-dependent amidation reaction. However, the exact molecular role of Dph7 is unclear. Here we demonstrate that Dph7 is an enzyme catalyzing a previously unknown step in the diphthamide biosynthesis pathway. This step is between the Dph5- and Dph6-catalyzed reactions. We demonstrate that the Dph5-catalyzed reaction generates methylated diphthine, a previously overlooked intermediate, and Dph7 is a methylesterase that hydrolyzes methylated diphthine to produce diphthine and allows the Dph6-catalyzed amidation reaction to occur. Thus, our study characterizes the molecular function of Dph7 for the first time and provides a revised diphthamide biosynthesis pathway.
- Published
- 2014
28. Glutamine Anabolism Plays a Critical Role in Pancreatic Cancer by Coupling Carbon and Nitrogen Metabolism
- Author
-
Jennifer P. Morton, David A. Tuveson, Vrushank Bhatt, Ya Ping Jiang, Howard C. Crawford, Eric Chiles, Wei-Xing Zong, Hua Zhong, Richard Z. Lin, Lanjing Zhang, Wouter H. Lamers, Hui Feng, Jessie Yanxiang Guo, Stephani Velasquez, Alex J. Bott, Eileen White, Jian Jin, Tracy G. Anthony, Nathiya Muthalagu, Nithya Sivaram, Xufen Yu, Jianliang Shen, Xiaoyang Su, Daniel J. Murphy, Ji-An Pan, Weiwei Dai, Claudia Tonelli, Sara Maimouni, Le Zhan, Tytgat Institute for Liver and Intestinal Research, and AGEM - Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
Male ,0301 basic medicine ,Anabolism ,endocrine system diseases ,Nitrogen ,Glutamine ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Glutamate-Ammonia Ligase ,Cell Line, Tumor ,Glutamine synthetase ,Pancreatic cancer ,medicine ,Animals ,Humans ,lcsh:QH301-705.5 ,Cell Proliferation ,Glutaminolysis ,Chemistry ,Metabolism ,medicine.disease ,Carbon ,3. Good health ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,Citric acid cycle ,030104 developmental biology ,lcsh:Biology (General) ,Cancer cell ,Cancer research ,Ketoglutaric Acids ,Female ,Gene Deletion ,030217 neurology & neurosurgery ,Carcinoma, Pancreatic Ductal - Abstract
SUMMARY Glutamine is thought to play an important role in cancer cells by being deaminated via glutaminolysis to α-ketoglutarate (aKG) to fuel the tricarboxylic acid (TCA) cycle. Supporting this notion, aKG supplementation can restore growth/survival of glutamine-deprived cells. However, pancreatic cancers are often poorly vascularized and limited in glutamine supply, in alignment with recent concerns on the significance of glutaminolysis in pancreatic cancer. Here, we show that aKG-mediated rescue of glutamine-deprived pancreatic ductal carcinoma (PDAC) cells requires glutamate ammonia ligase (GLUL), the enzyme responsible for de novo glutamine synthesis. GLUL-deficient PDAC cells are capable of the TCA cycle but defective in aKG-coupled glutamine biosynthesis and subsequent nitrogen anabolic processes. Importantly, GLUL expression is elevated in pancreatic cancer patient samples and in mouse PDAC models. GLUL ablation suppresses the development of KrasG12D-driven murine PDAC. Therefore, GLUL-mediated glutamine biosynthesis couples the TCA cycle with nitrogen anabolism and plays a critical role in PDAC., In Brief Bott et al. demonstrate that GLUL-mediated glutamine synthesis plays a critical role in converging the TCA cycle and nitrogen metabolism to promote nitrogen-dependent anabolic processes in pancreatic cancer. Ablation of GLUL suppresses PDAC development and may have important clinical implications., Graphical Abstract
- Published
- 2019
29. Ultrasensitive All-Carbon Photoelectrochemical Bioprobes for Zeptomole Immunosensing of Tumor Markers by an Inexpensive Visible Laser Light
- Author
-
Juan Wang, Shengshui Hu, Wangze Wu, Chengguo Hu, Xiaoyang Su, and Jinou Zheng
- Subjects
Immunoassay ,Detection limit ,Chromatography ,Green laser light ,biology ,Photochemistry ,Chemistry ,Lasers ,Visible laser light ,Analytical chemistry ,chemistry.chemical_element ,Covalent binding ,Electrochemical Techniques ,Serum samples ,Carbon ,Analytical Chemistry ,Carcinoembryonic antigen ,Microscopy, Electron, Transmission ,Limit of Detection ,Molecular Probes ,Biomarkers, Tumor ,biology.protein ,Humans ,Visible spectrum - Abstract
A novel enzyme-free and all-carbon photoelectrochemical (PEC) bioprobe, based on carboxylated multiwalled carbon nanotube-Congo red-fullerene nanohybrids (MWNTCOOH-CR-C60), for the ultrasensitive immunosensing of carcinoembryonic antigen (CEA) was reported. The MWNTCOOH-CR-C60 nanohybrids, prepared by mechanically grinding a mixture of MWNTCOOH, C60, and CR at a certain mass ratio, had good water dispersibility and high PEC conversion efficiency in visible light ranges. Covalent binding of the detection antibody of CEA on the MWNTCOOH-CR-C60 nanohybrids produced a sensitive PEC bioprobe for detection of CEA by sandwich immunosensing. The corresponding immunosensor, employing an inexpensive and portable green laser light, possessed a wide calibration range of 1.0 pg/mL~100.0 ng/mL and a low detection limit of 0.1 pg/mL (calculated 5 zmol for a 10.0 μL sample solution) (S/N = 3), which was successfully applied to the detection of CEA in serum samples from both healthy people and cancer patients. The present work thus demonstrated the promising application of fullerene-based nanocomposites in developing highly sensitive, environmentally friendly, and cost-effective PEC biosensors.
- Published
- 2013
30. Chemogenomic approach identified yeast YLR143W as diphthamide synthetase
- Author
-
Sheng Zhang, Wei Chen, Zhewang Lin, Xiaoyang Su, Hening Lin, and Hong Jiang
- Subjects
Saccharomyces cerevisiae Proteins ,Molecular Sequence Data ,Saccharomyces cerevisiae ,Genome ,Ligases ,chemistry.chemical_compound ,Plasmid ,Species Specificity ,Biosynthesis ,Tandem Mass Spectrometry ,Rosaniline Dyes ,Chemogenomics ,Humans ,Carbon-Nitrogen Ligases ,Histidine ,Amino Acid Sequence ,Cloning, Molecular ,Gene ,DNA Primers ,Genetics ,Multidisciplinary ,Molecular Structure ,biology ,Diphthamide ,Genomics ,Sequence Analysis, DNA ,Biological Sciences ,biology.organism_classification ,Yeast ,Biosynthetic Pathways ,chemistry ,Autoradiography ,Electrophoresis, Polyacrylamide Gel ,Phosphorus Radioisotopes ,Chromatography, Liquid ,Plasmids - Abstract
Many genes are of unknown functions in any sequenced genome. A combination of chemical and genetic perturbations has been used to investigate gene functions. Here we present a case that such “chemogenomics” information can be effectively used to identify missing genes in a defined biological pathway. In particular, we identified the previously unknown enzyme diphthamide synthetase for the last step of diphthamide biosynthesis. We found that yeast protein YLR143W is the diphthamide synthetase catalyzing the last amidation step using ammonium and ATP. Diphthamide synthetase is evolutionarily conserved in eukaryotes. The previously uncharacterized human gene ATPBD4 is the ortholog of yeast YLR143W and fully rescues the deletion of YLR143W in yeast.
- Published
- 2012
31. Protein Lysine Acylation and Cysteine Succination by Intermediates of Energy Metabolism
- Author
-
Bin He, Hening Lin, and Xiaoyang Su
- Subjects
Acylation ,Lysine ,macromolecular substances ,Nicotinamide adenine dinucleotide ,Biology ,Biochemistry ,Article ,chemistry.chemical_compound ,Succinylation ,Animals ,Humans ,Cysteine ,Cysteine metabolism ,Myristoylation ,Proteins ,General Medicine ,chemistry ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,NAD+ kinase ,Energy Metabolism ,Protein Processing, Post-Translational - Abstract
In the last few years, several new protein post-translational modifications that use intermediates in metabolism have been discovered. These include various acyl lysine modifications (formylation, propionylation, butyrylation, crotonylation, malonylation, succinylation, myristoylation) and cysteine succination. Here, we review the discovery and the current understanding of these modifications. Several of these modifications are regulated by the deacylases, sirtuins, which use nicotinamide adenine dinucleotide (NAD), an important metabolic small molecule. Interestingly, several of these modifications in turn regulate the activity of metabolic enzymes. These new modifications reveal interesting connections between metabolism and protein post-translational modifications and raise many questions for future investigations.
- Published
- 2012
32. Metabolic control of methylation and acetylation
- Author
-
Kathryn E. Wellen, Joshua D. Rabinowitz, and Xiaoyang Su
- Subjects
0301 basic medicine ,Biology ,Biochemistry ,Methylation ,Microbiology ,Article ,Analytical Chemistry ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,Animals ,Humans ,Epigenetics ,chemistry.chemical_classification ,Acetylation ,DNA ,030104 developmental biology ,Enzyme ,Histone ,chemistry ,Metabolic control analysis ,biology.protein ,NAD+ kinase - Abstract
Methylation and acetylation of DNA and histone proteins are the chemical basis for epigenetics. From bacteria to humans, methylation and acetylation are sensitive to cellular metabolic status. Modification rates depend on the availability of one-carbon and two-carbon substrates (S-adenosylmethionine, acetyl-CoA, and in bacteria also acetyl-phosphate). In addition, they are sensitive to demodification enzyme cofactors (α-ketoglutarate, NAD(+)) and structural analog metabolites that function as epigenetic enzyme inhibitors (e.g., S-adenosylhomocysteine, 2-hydroxyglutarate). Methylation and acetylation likely initially evolved to tailor protein activities in microbes to their metabolic milieu. While the extracellular environment of mammals is more tightly controlled, the combined impact of nutrient abundance and metabolic enzyme expression impacts epigenetics in mammals sufficiently to drive important biological outcomes such as stem cell fate and cancer.
- Published
- 2015
33. Detecting sirtuin-catalyzed deacylation reactions using ³²P-labeled NAD and thin-layer chromatography
- Author
-
Anita, Zhu, Xiaoyang, Su, and Hening, Lin
- Subjects
Kinetics ,Glutamate Dehydrogenase ,Lysine ,Humans ,Sirtuins ,Acetylation ,Chromatography, Thin Layer ,NAD ,Protein Processing, Post-Translational ,Peptide Fragments ,Phosphates ,Substrate Specificity - Abstract
Sirtuins are a class of enzymes with important functions in regulation aging, metabolism, and genome stability. They were originally known as nicotinamide adenine dinucleotide (NAD)-dependent protein lysine deacetylases. However, recently it has been discovered that certain sirtuins with weak deacetylase activity also hydrolyze novel acyl lysine modifications. These findings indicate that other sirtuins with weak deacetylase activity may also possess novel activities on unknown protein posttranslational modifications. Analytical methods that can help to identify new activity of sirtuins and new acyl lysine modifications are thus needed. Here we describe a sensitive method that uses (32)P-labeled NAD and thin-layer chromatography to detect sirtuin-catalyzed deacylation reactions. This method can help to discover new acyl lysine modifications that can be removed by novel sirtuin activities.
- Published
- 2013
34. Sirt5 Is an NAD-Dependent Protein Lysine Demalonylase and Desuccinylase
- Author
-
Johan Auwerx, Quan Hao, Wei Chen, Hening Lin, Yeyun Zhou, Richard A. Cerione, Bin He, Jungwoo Kim, Jun Huyn Kim, Jintang Du, Jiujiu Yu, Brian Hyun Choi, Saba Khan, Sheng Zhang, Jimin Woo, Hong Jiang, and Xiaoyang Su
- Subjects
Male ,Lysine ,Succinic Acid ,Mitochondria, Liver ,Nicotinamide adenine dinucleotide ,Crystallography, X-Ray ,Malonyl-Coa ,Histones ,Protein succinylation ,Succinylation ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Sirtuins ,Enzyme activity ,Inhibition ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,biology ,Deacetylases ,Acetylation ,Biochemistry ,030220 oncology & carcinogenesis ,Sirtuin ,Peptide ,Mechanism ,Hydrophobic and Hydrophilic Interactions ,Deacetylase activity ,SIRT5 ,Cells ,Carbamoyl-Phosphate Synthase (Ammonia) ,Insights ,Article ,03 medical and health sciences ,Animals ,Humans ,030304 developmental biology ,Mass spectrometry ,organic chemicals ,Hydrogen Bonding ,Structural Basis ,NAD ,Phosphorus 32 ,Kinetics ,chemistry ,biology.protein ,bacteria ,Cattle ,NAD+ kinase ,Peptides ,Protein Processing, Post-Translational - Abstract
Silent information regulator 2 (Sir2) proteins (sirtuins) are nicotinamide adenine dinucleotide-dependent deacetylases that regulate important biological processes. Mammals have seven sirtuins, Sirt1 to Sirt7. Four of them (Sirt4 to Sirt7) have no detectable or very weak deacetylase activity. We found that Sirt5 is an efficient protein lysine desuccinylase and demalonylase in vitro. The preference for succinyl and malonyl groups was explained by the presence of an arginine residue (Arg 105) and tyrosine residue (Tyr 102) in the acyl pocket of Sirt5. Several mammalian proteins were identified with mass spectrometry to have succinyl or malonyl lysine modifications. Deletion of Sirt5 in mice appeared to increase the level of succinylation on carbamoyl phosphate synthase 1, which is a known target of Sirt5. Thus, protein lysine succinylation may represent a posttranslational modification that can be reversed by Sirt5 in vivo., postprint
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.