Chrebp Regulates Fructose-Induced Glucose Production Independently of Insulin Signaling

Total Page:16

File Type:pdf, Size:1020Kb

Chrebp Regulates Fructose-Induced Glucose Production Independently of Insulin Signaling ChREBP regulates fructose-induced glucose production independently of insulin signaling Mi-Sung Kim, … , Michelle Lai, Mark A. Herman J Clin Invest. 2016;126(11):4372-4386. https://doi.org/10.1172/JCI81993. Research Article Endocrinology Metabolism Obese, insulin-resistant states are characterized by a paradoxical pathogenic condition in which the liver appears to be selectively insulin resistant. Specifically, insulin fails to suppress glucose production, yet successfully stimulates de novo lipogenesis. The mechanisms underlying this dysregulation remain controversial. Here, we hypothesized that carbohydrate-responsive element-binding protein (ChREBP), a transcriptional activator of glycolytic and lipogenic genes, plays a central role in this paradox. Administration of fructose increased hepatic hexose-phosphate levels, activated ChREBP, and caused glucose intolerance, hyperinsulinemia, hypertriglyceridemia, and hepatic steatosis in mice. Activation of ChREBP was required for the increased expression of glycolytic and lipogenic genes as well as glucose-6- phosphatase (G6pc) that was associated with the effects of fructose administration. We found that fructose-induced G6PC activity is a major determinant of hepatic glucose production and reduces hepatic glucose-6-phosphate levels to complete a homeostatic loop. Moreover, fructose activated ChREBP and induced G6pc in the absence of Foxo1a, indicating that carbohydrate-induced activation of ChREBP and G6PC dominates over the suppressive effects of insulin to enhance glucose production. This ChREBP/G6PC signaling axis is conserved in humans. Together, these findings support a carbohydrate-mediated, ChREBP-driven mechanism that contributes to hepatic insulin resistance. Find the latest version: https://jci.me/81993/pdf RESEARCH ARTICLE The Journal of Clinical Investigation ChREBP regulates fructose-induced glucose production independently of insulin signaling Mi-Sung Kim,1 Sarah A. Krawczyk,1 Ludivine Doridot,1 Alan J. Fowler,1 Jennifer X. Wang,2 Sunia A. Trauger,2 Hye-Lim Noh,3 Hee Joon Kang,3 John K. Meissen,4 Matthew Blatnik,4 Jason K. Kim,3 Michelle Lai,5 and Mark A. Herman1,6 1Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA. 2Faculty of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA. 3Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA. 4Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, Groton, Connecticut, USA. 5Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA. 6Broad Institute, Cambridge, Massachusetts, USA. Obese, insulin-resistant states are characterized by a paradoxical pathogenic condition in which the liver appears to be selectively insulin resistant. Specifically, insulin fails to suppress glucose production, yet successfully stimulates de novo lipogenesis. The mechanisms underlying this dysregulation remain controversial. Here, we hypothesized that carbohydrate- responsive element-binding protein (ChREBP), a transcriptional activator of glycolytic and lipogenic genes, plays a central role in this paradox. Administration of fructose increased hepatic hexose-phosphate levels, activated ChREBP, and caused glucose intolerance, hyperinsulinemia, hypertriglyceridemia, and hepatic steatosis in mice. Activation of ChREBP was required for the increased expression of glycolytic and lipogenic genes as well as glucose-6-phosphatase (G6pc) that was associated with the effects of fructose administration. We found that fructose-induced G6PC activity is a major determinant of hepatic glucose production and reduces hepatic glucose-6-phosphate levels to complete a homeostatic loop. Moreover, fructose activated ChREBP and induced G6pc in the absence of Foxo1a, indicating that carbohydrate-induced activation of ChREBP and G6PC dominates over the suppressive effects of insulin to enhance glucose production. This ChREBP/G6PC signaling axis is conserved in humans. Together, these findings support a carbohydrate-mediated, ChREBP-driven mechanism that contributes to hepatic insulin resistance. Introduction tor, proximal to the proposed branch points in insulin signaling, The metabolic syndrome is a cluster of disorders including obe- thereby precluding differential insulin signaling as a mechanism sity, nonalcoholic fatty liver disease (NAFLD), hypertriglyceri- to explain this paradox (7). It has also been suggested that insulin’s demia, and insulin resistance, which predisposes to the devel- acute effect of reducing hepatic glucose production (HGP) is inde- opment of type 2 diabetes and cardiovascular disease (1). This pendent of hepatic insulin signaling altogether and instead depends syndrome is also characterized by a pathogenic paradox in which on suppression of adipose lipolysis and delivery of fatty-acid sub- the liver appears selectively insulin resistant: hyperinsulinemia is strate to the liver (8, 9). However, this contrasts with other studies unable to appropriately suppress glucose production, but the liver indicating that insulin’s acute effect of reducing glucose production remains sensitive to insulin’s ability to stimulate de novo lipogen- is mediated through insulin’s actions directly on the liver (10, 11). esis (DNL) (2). These studies do not address the observation that hyperinsulinemia Despite extensive investigation, the mechanisms accounting fails to suppress hepatic glucose-6-phosphatase (G6PC) expression for this apparent paradox remain controversial. Investigation at in insulin-resistant humans (12), a condition where increased DNL the level of hepatic insulin signaling suggested that different arms in association with steatosis and hypertriglyceridemia is readily of the insulin signaling cascade might be differentially sensitive apparent (13, 14). Understanding the signaling mechanism regulat- to insulin (3–6). For instance, insulin signaling to inhibit the tran- ing G6pc in this context is important, as increased hepatic G6PC is scription factor forkhead box O1a (FOXO1A), which transactivates sufficient to cause glucose intolerance and insulin resistance (15). gluconeogenic enzyme gene expression, might be impaired, while Another mechanism that may contribute to the paradox is insulin-mediated stimulation of a mechanistic target of rapamycin that glucose or gluconeogenic substrate delivered to the liver can (MTOR)/sterol regulatory element–binding transcription factor 1 activate hepatic DNL independently of insulin signaling (16–19). (SREBPF1 or SREBP1) signaling axis, which upregulates a DNL gene Carbohydrate-responsive element-binding protein (ChREBP, also program, remains intact (5). However, others reported that defects known as MLXIPL) is a master transcriptional regulator of glyco- in hepatic insulin signaling occur at the level of the insulin recep- lytic and lipogenic genes and is activated by carbohydrate (CHO) metabolites in key metabolic tissues including liver. In insulin-resis- tant states, when glucose disposal is impaired in peripheral tissues Authorship note: M.S. Kim and S.A. Krawczyk contributed equally to this work. such as skeletal muscle and adipose tissue, shunting of glucose to Conflict of interest: J.K. Meissen and M. Blatnik are employees of Pfizer Inc. Submitted: March 23, 2015; Accepted: August 18, 2016. the liver might activate ChREBP and enhance DNL. For instance, Reference information: J Clin Invest. 2016;126(11):4372–4386. doi:10.1172/JCI81993. genetically ablating Glut4, the insulin-responsive glucose transport- 4372 jci.org Volume 126 Number 11 November 2016 The Journal of Clinical Investigation RESEARCH ARTICLE Figure 1. High-fructose feeding induces metabolic disease. (A) Body weight and (B) fat pad weights (PG, perigonadal; SC, subcutaneous; BAT, brown adipose tissue) were measured in WT male mice fed chow, high-dextrose, or high-fructose diet for 9 weeks (n = 8 per group). (C) Glycemia and (D) serum insulin were measured in the ad libitum–fed state. (E) Serum triglyceride (TG) levels were measured after an overnight fast followed by 3 hours refeeding with chow, glucose, or fructose diet. (F) Hepatic TG levels and representative H&E-stained liver sections. Images were obtained at ×20 magnification. (G) Glucose tolerance test and (H) glycerol tolerance test with incremental areas under the curve (Δ AUC) for this cohort. P values were obtained by 1-way ANOVA. *P < 0.05 versus chow; **P < 0.05 versus all others. Values are the mean ± SEM. er, from skeletal muscle and adipose tissue enhances hepatic DNL It has recently been hypothesized that hepatocyte CHO (20). Additionally, siRNA-mediated knockdown of ChREBP in metabolites (hexose- or triose-phosphates) might not only stimu- ob/ob mice decreases hepatic DNL in the setting of persistent hyper- late ChREBP and activate DNL, but might also serve as a signal insulinemia (21). Thus, hepatic DNL may be regulated by increased to enhance HGP (17, 18). This hypothesis derives in part from the substrate delivery independently of insulin signaling. However, counterintuitive observation that while ChREBP is known to stim- whether increasing intrahepatic CHO metabolites might also signal ulate glycolysis through transactivation of glycolytic genes (22), it to increase glucose production has not been fully explored. may also transactivate expression of G6pc encoding the enzyme jci.org Volume 126 Number 11 November 2016 4373 RESEARCH ARTICLE The Journal of Clinical Investigation
Recommended publications
  • Phenotypic and Molecular Genetic Analysis of Pyruvate Kinase Deficiency in a Tunisian Family
    The Egyptian Journal of Medical Human Genetics (2016) 17, 265–270 HOSTED BY Ain Shams University The Egyptian Journal of Medical Human Genetics www.ejmhg.eg.net www.sciencedirect.com ORIGINAL ARTICLE Phenotypic and molecular genetic analysis of Pyruvate Kinase deficiency in a Tunisian family Jaouani Mouna a,1,*, Hamdi Nadia a,1, Chaouch Leila a, Kalai Miniar a, Mellouli Fethi b, Darragi Imen a, Boudriga Imen a, Chaouachi Dorra a, Bejaoui Mohamed b, Abbes Salem a a Laboratory of Molecular and Cellular Hematology, Pasteur Institute, Tunis, Tunisia b Service d’Immuno-He´matologie Pe´diatrique, Centre National de Greffe de Moelle Osseuse, Tunis, Tunisia Received 9 July 2015; accepted 6 September 2015 Available online 26 September 2015 KEYWORDS Abstract Pyruvate Kinase (PK) deficiency is the most frequent red cell enzymatic defect responsi- Pyruvate Kinase deficiency; ble for hereditary non-spherocytic hemolytic anemia. The disease has been studied in several ethnic Phenotypic and molecular groups. However, it is yet an unknown pathology in Tunisia. We report here, the phenotypic and investigation; molecular investigation of PK deficiency in a Tunisian family. Hemolytic anemia; This study was carried out on two Tunisian brothers and members of their family. Hematolog- Hydrops fetalis; ical, biochemical analysis and erythrocyte PK activity were performed. The molecular characteriza- PKLR mutation tion was carried out by gene sequencing technique. The first patient died few hours after birth by hydrops fetalis, the second one presented with neonatal jaundice and severe anemia necessitating urgent blood transfusion. This severe clinical pic- ture is the result of a homozygous mutation of PKLR gene at exon 8 (c.1079G>A; p.Cys360Tyr).
    [Show full text]
  • Reduced Expression of Pyruvate Kinase in Kidney Proximal Tubule
    www.nature.com/scientificreports OPEN Reduced expression of pyruvate kinase in kidney proximal tubule cells is a potential mechanism Received: 29 March 2018 Accepted: 22 January 2019 of pravastatin altered glucose Published: xx xx xxxx metabolism Yong Pyo Lee1, Yuri Cho2, Eun Jee Kim2,3, Hyojung Lee2, Hoon Young Choi4, Hye Jin Wang5, Eun Seok Kang 5, Yu Seun Kim2,6, Myoung Soo Kim1,2,6 & Beom Seok Kim2,3,7 Recent studies have reported that statins are associated with increased incidence of diabetes. Although several mechanisms have been proposed, the role of the kidney’s glucose metabolism upon statin treatment is still unclear. Thus, we investigated the role of pravastatin in gluconeogenesis and glycolysis. HK-2 and HepG2 cells were treated with pravastatin and cultured under either high- or normal-cholesterol conditions. In HK-2 cells treated with pravastatin under both high- and normal- cholesterol conditions, the protein expression of only pyruvate kinase isozymes L/R (PKLR) decreased in a dose-dependent manner, while the protein expression of other glucose metabolism related enzymes remained unchanged. Within the in vivo experiment, male C57BL/6 mice were fed either pravastatin-treated normal-fat diets for 2 or 4 weeks or pravastatin-treated high-fat diets for 16 weeks. Protein expression of PKLR in the kidneys from mice that consumed pravastatin-treated high-fat diets decreased signifcantly compared to the controls. Upon the treatments of pravastatin, only the PKLR expression decreased in lean mice. Furthermore, PKLR activity decreased signifcantly in the kidney after pravastatin treatments. However, there was no change in enzyme activity in the liver, suggesting that pravastatin decreased PKLR activity only in the kidney.
    [Show full text]
  • Supporting Information
    Supporting Information Wakabayashi et al. 10.1073/pnas.1521754113 SI Materials and Methods DNA Transfection and Puromycin Selection. K562 cells were cotrans- Plasmid Preparation. Short guide RNA (sgRNA) sequences (Table fected with 1 μg total of Cas9 nuclease and sgRNA plasmids using S1) were cloned into the pSg1 vector (Addgene) and the XPR5 Lipofectamine LTX Plus Reagent (Thermo Fisher Scientific) at a lentiviral vector (Broad Institute), respectively. [The XPR5 1:2 ratio of Cas9 to sgRNA. For a control, K562 cells were co- vector contains the Cas9 nuclease and a red fluorescent protein transfectedwith1μg total of Cas9 nuclease and pLKO.1-GFP (RFP) cassette.] The Cas9 nuclease expression vector used was plasmid at a 1:2 ratio of Cas9 to pLKO.1-GFP. At 24 h after co- μ pxPR_BRD001, which contains a puromycin resistance cassette transfection, puromycin was added at a concentration of 2 g/mL, μ as a selection marker. Off-target scores for each guide were followed 24 h later by a reduction to 1 g/mL for an additional 24 h. calculated using the CRISPR design tool (CRISPR Design; Selection efficiency was assessed by flow cytometry with propidium crispr.mit.edu); only guides with a score >50 (except for a score iodide staining (to assess viability) on a FACSCanto II flow cy- of 49 in one case) were used. tometer (BD Biosciences). Limiting dilutions were performed to obtain single cell-derived clonal populations for both cells targeted Cell Culture and Lentivirus Production. The K562 cells (American with sgRNAs as well as for GFP controls. Unless specified other- Type Culture Collection) were maintained in RPMI medium 1640 wise, three matching clonal GFP controls were analyzed for each plus L-glutamine (Life Technologies) supplemented with 10% experiment.
    [Show full text]
  • Protein Interactions in the Cancer Proteome† Cite This: Mol
    Molecular BioSystems View Article Online PAPER View Journal | View Issue Small-molecule binding sites to explore protein– protein interactions in the cancer proteome† Cite this: Mol. BioSyst., 2016, 12,3067 David Xu,ab Shadia I. Jalal,c George W. Sledge Jr.d and Samy O. Meroueh*aef The Cancer Genome Atlas (TCGA) offers an unprecedented opportunity to identify small-molecule binding sites on proteins with overexpressed mRNA levels that correlate with poor survival. Here, we analyze RNA-seq and clinical data for 10 tumor types to identify genes that are both overexpressed and correlate with patient survival. Protein products of these genes were scanned for binding sites that possess shape and physicochemical properties that can accommodate small-molecule probes or therapeutic agents (druggable). These binding sites were classified as enzyme active sites (ENZ), protein–protein interaction sites (PPI), or other sites whose function is unknown (OTH). Interestingly, the overwhelming majority of binding sites were classified as OTH. We find that ENZ, PPI, and OTH binding sites often occurred on the same structure suggesting that many of these OTH cavities can be used for allosteric modulation of Creative Commons Attribution 3.0 Unported Licence. enzyme activity or protein–protein interactions with small molecules. We discovered several ENZ (PYCR1, QPRT,andHSPA6)andPPI(CASC5, ZBTB32,andCSAD) binding sites on proteins that have been seldom explored in cancer. We also found proteins that have been extensively studied in cancer that have not been previously explored with small molecules that harbor ENZ (PKMYT1, STEAP3,andNNMT) and PPI (HNF4A, MEF2B,andCBX2) binding sites. All binding sites were classified by the signaling pathways to Received 29th March 2016, which the protein that harbors them belongs using KEGG.
    [Show full text]
  • Glucose Catabolism in Liver Tumors Induced by C-MYC Can Be
    Published OnlineFirst June 19, 2017; DOI: 10.1158/0008-5472.CAN-17-0498 Cancer Tumor and Stem Cell Biology Research Glucose Catabolism in Liver Tumors Induced by c-MYC Can Be Sustained by Various PKM1/PKM2 Ratios and Pyruvate Kinase Activities Andres Mendez-Lucas 1, Xiaolei Li2,3, Junjie Hu2,4, Li Che2, Xinhua Song2, Jiaoyuan Jia2,5, Jingxiao Wang2, Chencheng Xie2,6, Paul C. Driscoll1, Darjus F. Tschaharganeh7,8, Diego F. Calvisi9, Mariia Yuneva1, and Xin Chen2,4 Abstract Different pyruvate kinase isoforms are expressed in a tissue- tumorigenesis and resulted in liver tumor formation with specific manner, with pyruvate kinase M2 (PKM2) suggested to decreased pyruvate kinase activity and decreased catabolism be the predominant isoform in proliferating cells and cancer of glucose into alanine and the Krebs cycle. An increased PKM1/ cells. Because of differential regulation of enzymatic activities, PKM2 ratio by ectopic PKM1 expression further decreased PKM2, but not PKM1, has been thought to favor cell prolifer- glucose flux into serine biosynthesis and increased flux into ation. However, the role of PKM2 in tumorigenesis has been lactate and the Krebs cycle, resulting in reduced total levels of recently challenged. Here we report that increased glucose serine. However, these changes also did not affect c-MYC– catabolism through glycolysis and increased pyruvate kinase induced liver tumor development. These results suggest that activity in c-MYC-driven liver tumors are associated with increased expression of PKM2 is not required to support increased expression of both PKM1 and PKM2 isoforms and c-MYC–induced tumorigenesis in the liver and that various decreased expression of the liver-specific isoform of pyruvate PKM1/PKM2 ratios and pyruvate kinase activities can sustain kinase, PKL.
    [Show full text]
  • Chrebp-Knockout Mice Show Sucrose Intolerance and Fructose Malabsorption
    nutrients Article ChREBP-Knockout Mice Show Sucrose Intolerance and Fructose Malabsorption Takehiro Kato 1, Katsumi Iizuka 1,2,* ID , Ken Takao 1, Yukio Horikawa 1, Tadahiro Kitamura 3 and Jun Takeda 1 1 Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; [email protected] (T.K.); [email protected] (K.T.); [email protected] (Y.H.); [email protected] (J.T.) 2 Gifu University Hospital Center for Nutritional Support and Infection Control, Gifu 501-1194, Japan 3 Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; [email protected] * Correspondence: [email protected]; Tel.: +81-58-230-6564; Fax: +81-58-230-6376 Received: 31 January 2018; Accepted: 9 March 2018; Published: 10 March 2018 Abstract: We have previously reported that 60% sucrose diet-fed ChREBP knockout mice (KO) showed body weight loss resulting in lethality. We aimed to elucidate whether sucrose and fructose metabolism are impaired in KO. Wild-type mice (WT) and KO were fed a diet containing 30% sucrose with/without 0.08% miglitol, an α-glucosidase inhibitor, and these effects on phenotypes were tested. Furthermore, we compared metabolic changes of oral and peritoneal fructose injection. A thirty percent sucrose diet feeding did not affect phenotypes in KO. However, miglitol induced lethality in 30% sucrose-fed KO. Thirty percent sucrose plus miglitol diet-fed KO showed increased cecal contents, increased fecal lactate contents, increased growth of lactobacillales and Bifidobacterium and decreased growth of clostridium cluster XIVa.
    [Show full text]
  • Network Modeling Approaches and Applications to Unravelling Non-Alcoholic Fatty Liver Disease
    G C A T T A C G G C A T genes Review Network Modeling Approaches and Applications to Unravelling Non-Alcoholic Fatty Liver Disease Montgomery Blencowe 1,2 , Tilan Karunanayake 1, Julian Wier 1, Neil Hsu 1 and Xia Yang 1,2,3,* 1 Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA; [email protected] (M.B.); [email protected] (T.K.); [email protected] (J.W.); [email protected] (N.H.) 2 Molecular, Cellular, and Integrative Physiology Interdepartmental Program, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA 3 Interdepartmental Program of Bioinformatics, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, USA * Correspondence: [email protected] Received: 22 October 2019; Accepted: 22 November 2019; Published: 24 November 2019 Abstract: Non-alcoholic fatty liver disease (NAFLD) is a progressive condition of the liver encompassing a range of pathologies including steatosis, non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma. Research into this disease is imperative due to its rapid growth in prevalence, economic burden, and current lack of FDA approved therapies. NAFLD involves a highly complex etiology that calls for multi-tissue multi-omics network approaches to uncover the pathogenic genes and processes, diagnostic biomarkers, and potential therapeutic strategies. In this review, we first present a basic overview of disease pathogenesis, risk factors, and remaining knowledge gaps, followed by discussions of the need and concepts of multi-tissue multi-omics approaches, various network methodologies and application examples in NAFLD research.
    [Show full text]
  • Metabolic Network-Based Stratification of Hepatocellular Carcinoma Reveals Three Distinct Tumor Subtypes
    Metabolic network-based stratification of hepatocellular carcinoma reveals three distinct tumor subtypes Gholamreza Bidkhoria,b,1, Rui Benfeitasa,1, Martina Klevstigc,d, Cheng Zhanga, Jens Nielsene, Mathias Uhlena, Jan Borenc,d, and Adil Mardinoglua,b,e,2 aScience for Life Laboratory, KTH Royal Institute of Technology, SE-17121 Stockholm, Sweden; bCentre for Host-Microbiome Interactions, Dental Institute, King’s College London, SE1 9RT London, United Kingdom; cDepartment of Molecular and Clinical Medicine, University of Gothenburg, SE-41345 Gothenburg, Sweden; dThe Wallenberg Laboratory, Sahlgrenska University Hospital, SE-41345 Gothenburg, Sweden; and eDepartment of Biology and Biological Engineering, Chalmers University of Technology, SE-41296 Gothenburg, Sweden Edited by Sang Yup Lee, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea, and approved November 1, 2018 (received for review April 27, 2018) Hepatocellular carcinoma (HCC) is one of the most frequent forms of of markers associated with recurrence and poor prognosis (13–15). liver cancer, and effective treatment methods are limited due to Moreover, genome-scale metabolic models (GEMs), collections tumor heterogeneity. There is a great need for comprehensive of biochemical reactions, and associated enzymes and transporters approaches to stratify HCC patients, gain biological insights into have been successfully used to characterize the metabolism of subtypes, and ultimately identify effective therapeutic targets. We HCC, as well as identify drug targets for HCC patients (11, 16–18). stratified HCC patients and characterized each subtype using tran- For instance, HCC tumors have been stratified based on the uti- scriptomics data, genome-scale metabolic networks and network lization of acetate (11). Analysis of HCC metabolism has also led topology/controllability analysis.
    [Show full text]
  • Download Download
    Journal of Pediatric Academy Year: 2021 Volume: 2 Issue: 1 Invited Review Doi: 10.51271/jpea-2021-0120 Hereditary Red Blood Cell Enzymopathies Author(s) Tekin Aksu Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Hematology, Ankara, Affiliation(s) Turkey Article Type: Invited Review Received: 15.03.2021 Article Article Group: Pediatric Hematology Accepted: 21.04.2021 Information Available Online: 30.04.2021 Cite this article as: Aksu T. Hereditary Red Blood Cell Enzymopathies. J Pediatr Acad 2021; 2: 9-13. Abstract Red cell metabolic disturbances result in hemolysis, which leads to a significant shortening of the erythrocyte life span. The most common enzyme deficiencies are glucose 6-phosphate dehydrogenase (G6PD) in the antioxidant pathway, pyruvate kinase in the anaerobic glycolysis pathway, and pyrimidine 5’ nucleotidase (P5’N) in the nucleotide metabolism. While the X chromosome inherits G6PD and phosphoglycerate kinase deficiencies, other enzymopathies show autosomal recessive inheritance. Although the causes of hereditary hemolytic disorders are diverse, clinical, laboratory findings and complications overlap. A history of neonatal jaundice requiring phototherapy and exchange transfusion is quite usual. Mild to severe anemia may be accompanied with episodic or constant hemolysis associated with icterus, hyperbilirubinemia, growth retardation, gallstones, splenomegaly, and a variable degree of iron overload. Erythrocyte enzyme disorders should be suspected in patients with severe hemolytic episodes, or chronic hemolysis, after excluding hemoglobinopathies, membranopathies, and immune-mediated hemolysis. Keywords: Glucose 6-phosphate dehydrogenase deficiency, pyruvate kinase deficiency, glucose phosphate isomerase deficiency, triosephosphate isomerase deficiency, pyrimidine 5’-nucleotidase deficiency Introduction Erythrocytes use two main biochemical pathways to maintain The most common enzyme deficiencies are glucose their physiological state.
    [Show full text]
  • Non-Commercial Use Only
    Thalassemia Reports 2013; volume 3(s1):e33 Diagnosis and epidemiology of red blood cell enzyme disorders Richard Van Wijk Associate professor, Laboratory For Red Blood Cell Research, Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, The Netherlands The red blood cell possess an active metabolic machinery that pro- tion of fava beans. Patients with uncommon, functionally very severe, vides the cell with energy to pump ions against electrochemical gradi- genetic variants of G6PD experience chronic hemolysis, a disorder ents, to maintain its shape, to keep hemoglobin iron in the reduced designated hereditary nonspherocytic hemolytic anemia (HNSHA).(3) (ferrous) form, and to maintain enzyme and hemoglobin sulfhydryl Hereditary nonspherocytic hemolytic anemia also occurs as a conse- groups. The main source of metabolic energy comes from glucose. quence of other enzyme deficiencies, the most common of which is Glucose is metabolized through the glycolytic pathway and through the pyruvate kinase (PK) deficiency.(4-6) Deficiencies of glucosephos- hexose monophosphate shunt. Glycolysis catabolizes glucose to pyru- phate isomerase(6), triosephosphate isomerase(7), and pyrimidine 52 vate and lactate, which represent the end products of glucose metabo- -nucleotidase deficiency(8) are included among the relatively rare lism in the erythrocyte. Adenosine diphosphate (ADP) is phosphorylat- causes of HNSHA. In the case of some deficiencies, notably those of ed to adenosine triphosphate (ATP), and nicotinamide adenine dinu- glutathione synthetase(9), triosephosphate isomerase(7), and phos- cleotide (NAD)+ is reduced to NADH in glycolysis. 2,3- phoglycerate kinase(10), the defect is expressed throughout the body, Bisphosphoglycerate, an important regulator of the oxygen affinity of and neurologic and other defects may be a prominent part of the clin- hemoglobin, is generated during glycolysis by the Rapoport-Luebering ical syndrome.
    [Show full text]
  • Elevated Glucose Represses Liver Glucokinase and Induces Its Regulatory Protein to Safeguard Hepatic Phosphate Homeostasis Catherine Arden,1 John L
    ORIGINAL ARTICLE Elevated Glucose Represses Liver Glucokinase and Induces Its Regulatory Protein to Safeguard Hepatic Phosphate Homeostasis Catherine Arden,1 John L. Petrie,1 Susan J. Tudhope,1 Ziad Al-Oanzi,1,2 Amy J. Claydon,3 Robert J. Beynon,3 Howard C. Towle,4 and Loranne Agius1 OBJECTIVE—The induction of hepatic glucose 6-phosphatase in the concentrations of Gck and Gckr, as shown by the (G6pc) by glucose presents a paradox of glucose-induced glucose unusually high control strength of these proteins on glu- intolerance. We tested whether glucose regulation of liver gene cose metabolism (2–4). Consistent with the major role for expression is geared toward intracellular homeostasis. Gck in control of blood glucose homeostasis, transcription of the Gck gene in the liver is induced by insulin and re- RESEARCH DESIGN AND METHODS—The effect of glucose- pressed by glucagon (5). Type 2 diabetes is associated with induced accumulation of phosphorylated intermediates on ex- – pression of glucokinase (Gck) and its regulator Gckr was defects in hepatic glucose uptake and production (6 10), determined in hepatocytes. Cell ATP and uric acid production and a role for defective Gck regulation or expression has were measured as indices of cell phosphate homeostasis. been implicated (8–10); however the underlying mecha- — nisms are unresolved. RESULTS Accumulation of phosphorylated intermediates in Liver Gck is a major determinant of the hepatic con- hepatocytes incubated at elevated glucose induced rapid and inverse changes in Gck (repression) and Gckr (induction) mRNA centration of glucose 6-P, which is a linear function of the concomitantly with induction of G6pc, but had slower effects on free Gck activity (1).
    [Show full text]
  • Genetic Diversity in Human Erythrocyte Pyruvate Kinase
    Genes and Immunity (2012) 13, 98–102 & 2012 Macmillan Publishers Limited All rights reserved 1466-4879/12 www.nature.com/gene SHORT COMMUNICATION Genetic diversity in human erythrocyte pyruvate kinase J Berghout1, S Higgins2, C Loucoubar3, A Sakuntabhai3,4, KC Kain2 and P Gros1 1Department of Biochemistry, McGill University, Complex Traits Group, Montreal, Canada; 2McLaughlin-Rotman Centre for Global Health, Toronto General Hospital, University of Toronto, Toronto, Canada; 3Institut Pasteur, Unite´ de Pathoge´nie Virale, Paris, France and 4Center of Excellence for Vector and Vector-Borne Diseases, Faculty of Science, Mahidol University, Bangkok, Thailand Previously, we have shown that pyruvate kinase, liver and red cell isoform (PKLR) deficiency protects mice in vivo against blood-stage malaria, and observed that reduced PKLR function protects human erythrocytes against Plasmodium falciparum replication ex vivo. Here, we have sequenced the human PKLR gene in 387 individuals from malaria-endemic and other regions in order to assess genetic variability in different geographical regions and ethnic groups. Rich genetic diversity was detected in PKLR, including 59 single-nucleotide polymorphisms and several loss-of-function variants (frequency 1.5%). Haplotype distribution and allele frequency varied considerably with geography. Neutrality testing suggested positive selection of the genein the sub-Saharan African and Pakistan populations. It is possible that such positive selection involves the malarial parasite. Genes and Immunity (2012) 13, 98–102; doi:10.1038/gene.2011.54; published online 11 August 2011 Keywords: genetics; pyruvate kinase; sequencing; genetic diversity; CEPH; malaria Introduction polymorphisms (SNPs; Figure 1b, Supplementary Table 1 for details). Full nucleotide sequences can be found Erythrocytes rely exclusively on glycolysis for production by accessing the CEPH database.
    [Show full text]