1
|
Alturki MS. Exploring Marine-Derived Compounds: In Silico Discovery of Selective Ketohexokinase (KHK) Inhibitors for Metabolic Disease Therapy. Mar Drugs 2024; 22:455. [PMID: 39452863 PMCID: PMC11509851 DOI: 10.3390/md22100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
The increasing prevalence of metabolic diseases, including nonalcoholic fatty liver disease (NAFLD), obesity, and type 2 diabetes, poses significant global health challenges. Ketohexokinase (KHK), an enzyme crucial in fructose metabolism, is a potential therapeutic target due to its role in these conditions. This study focused on the discovery of selective KHK inhibitors using in silico methods. We employed structure-based drug design (SBDD) and ligand-based drug design (LBDD) approaches, beginning with molecular docking to identify promising compounds, followed by induced-fit docking (IFD), molecular mechanics generalized Born and surface area continuum solvation (MM-GBSA), and molecular dynamics (MD) simulations to validate binding affinities. Additionally, shape-based screening was conducted to assess structural similarities. The findings highlight several potential inhibitors with favorable ADMET profiles, offering promising candidates for further development in the treatment of fructose-related metabolic disorders.
Collapse
Affiliation(s)
- Mansour S Alturki
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
2
|
Ferreira JC, Villanueva AJ, Fadl S, Al Adem K, Cinviz ZN, Nedyalkova L, Cardoso THS, Andrade ME, Saksena NK, Sensoy O, Rabeh WM. Residues in the fructose-binding pocket are required for ketohexokinase-A activity. J Biol Chem 2024; 300:107538. [PMID: 38971308 PMCID: PMC11332825 DOI: 10.1016/j.jbc.2024.107538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024] Open
Abstract
Excessive fructose consumption is a primary contributor to the global surges in obesity, cancer, and metabolic syndrome. Fructolysis is not robustly regulated and is initiated by ketohexokinase (KHK). In this study, we determined the crystal structure of KHK-A, one of two human isozymes of KHK, in the apo-state at 1.85 Å resolution, and we investigated the roles of residues in the fructose-binding pocket by mutational analysis. Introducing alanine at D15, N42, or N45 inactivated KHK-A, whereas mutating R141 or K174 reduced activity and thermodynamic stability. Kinetic studies revealed that the R141A and K174A mutations reduced fructose affinity by 2- to 4-fold compared to WT KHK-A, without affecting ATP affinity. Molecular dynamics simulations provided mechanistic insights into the potential roles of the mutated residues in ligand coordination and the maintenance of an open state in one monomer and a closed state in the other. Protein-protein interactome analysis indicated distinct expression patterns and downregulation of partner proteins in different tumor tissues, warranting a reevaluation of KHK's role in cancer development and progression. The connections between different cancer genes and the KHK signaling pathway suggest that KHK is a potential target for preventing cancer metastasis. This study enhances our understanding of KHK-A's structure and function and offers valuable insights into potential targets for developing treatments for obesity, cancer, and metabolic syndrome.
Collapse
Affiliation(s)
- Juliana C Ferreira
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Adrian J Villanueva
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Samar Fadl
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Kenana Al Adem
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Zeynep Nur Cinviz
- Graduate School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul, Turkey
| | - Lyudmila Nedyalkova
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | | | - Mario Edson Andrade
- Horticultural Sciences Department, University of Florida, Gainesville, Florida, USA
| | - Nitin K Saksena
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Ozge Sensoy
- Graduate School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul, Turkey; Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Wael M Rabeh
- Science Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
3
|
Ortjohann M, Schönheit P. Identification and characterization of a novel type of ketohexokinase from the haloarchaeon Haloferax volcanii. FEMS Microbiol Lett 2024; 371:fnae026. [PMID: 38587824 DOI: 10.1093/femsle/fnae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/18/2024] [Accepted: 04/05/2024] [Indexed: 04/09/2024] Open
Abstract
Ketohexokinase (KHK) catalyzes the ATP-dependent phosphorylation of fructose, forming fructose-1-phosphate and ADP. The enzyme is well studied in Eukarya, in particular in humans and other vertebrates, but homologs have not been identified in Bacteria and Archaea. Here we report the identification of a novel type of KHK from the haloarchaeon Haloferax volcanii (HvKHK). The encoding gene khk was identified as HVO_1812. The gene was expressed as a 90-kDa homodimeric protein, catalyzing the phosphorylation of fructose with a Vmax value of 59 U/mg and apparent KM values for ATP and fructose of 0.47 and 1.29 mM, respectively. Homologs of HvKHK were only identified in a few haloarchaea and halophilic Bacteria. The protein showed low sequence identity to characterized KHKs from Eukarya and phylogenetic analyses indicate that haloarchaeal KHKs are largely separated from eukaryal KHKs. This is the first report of the identification of KHKs in prokaryotes that form a novel cluster of sugar kinases within the ribokinase/pfkB superfamily.
Collapse
Affiliation(s)
- Marius Ortjohann
- Institut für Allgemeine Mikrobiologie, Christian-Albrechts-Universität Kiel, Am Botanischen Garten 1-9, D-24118 Kiel, Germany
| | - Peter Schönheit
- Institut für Allgemeine Mikrobiologie, Christian-Albrechts-Universität Kiel, Am Botanischen Garten 1-9, D-24118 Kiel, Germany
| |
Collapse
|
4
|
Sakasai-Sakai A, Takeda K, Takeuchi M. Involvement of Intracellular TAGE and the TAGE-RAGE-ROS Axis in the Onset and Progression of NAFLD/NASH. Antioxidants (Basel) 2023; 12:antiox12030748. [PMID: 36978995 PMCID: PMC10045097 DOI: 10.3390/antiox12030748] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
The repeated excessive intake of sugar, a factor that contributes to the onset of nonalcoholic fatty liver disease (NAFLD) and its progression to the chronic form of nonalcoholic steatohepatitis (NASH), markedly increases the hepatocyte content of glyceraldehyde (GA), a glucose/fructose metabolic intermediate. Toxic advanced glycation end-products (toxic AGEs, TAGE) are synthesized by cross-linking reactions between the aldehyde group of GA and the amino group of proteins, and their accumulation has been implicated in the development of NAFLD/NASH and hepatocellular carcinoma (HCC). Our previous findings not only showed that hepatocyte disorders were induced by the intracellular accumulation of TAGE, but they also indicated that extracellular leakage resulted in elevated TAGE concentrations in circulating fluids. Interactions between extracellular TAGE and receptor for AGEs (RAGE) affect intracellular signaling and reactive oxygen species (ROS) production, which may, in turn, contribute to the pathological changes observed in NAFLD/NASH. RAGE plays a role in the effects of the extracellular leakage of TAGE on the surrounding cells, which ultimately promote the onset and progression of NAFLD/NASH. This review describes the relationships between intracellular TAGE levels and hepatocyte and hepatic stellate cell (HSC) damage as well as the TAGE-RAGE-ROS axis in hepatocytes, HSC, and HCC cells. The "TAGE theory" will provide novel insights for future research on NAFLD/NASH.
Collapse
Affiliation(s)
- Akiko Sakasai-Sakai
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Kenji Takeda
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan
| | - Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan
| |
Collapse
|
5
|
Takata T, Sakasai-Sakai A, Takeuchi M. Intracellular Toxic Advanced Glycation End-Products May Induce Cell Death and Suppress Cardiac Fibroblasts. Metabolites 2022; 12:metabo12070615. [PMID: 35888739 PMCID: PMC9321527 DOI: 10.3390/metabo12070615] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/18/2022] [Accepted: 06/23/2022] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease (CVD) is a lifestyle-related disease (LSRD) induced by the dysfunction and cell death of cardiomyocytes. Cardiac fibroblasts are activated and differentiate in response to specific signals, such as transforming growth factor-β released from injured cardiomyocytes, and are crucial for the protection of cardiomyocytes, cardiac tissue repair, and remodeling. In contrast, cardiac fibroblasts have been shown to induce injury or death of cardiomyocytes and are implicated in the pathogenesis of diseases such as cardiac hypertrophy. We designated glyceraldehyde-derived advanced glycation end-products (AGEs) as toxic AGEs (TAGE) due to their cytotoxicity and association with LSRD. Intracellular TAGE in cardiomyocytes decreased their beating rate and induced cell death in the absence of myocardial ischemia. The TAGE levels in blood were elevated in patients with CVD and were associated with myocardial ischemia along with increased risk of atherosclerosis in vascular endothelial cells in vitro. The relationships between the dysfunction or cell death of cardiac fibroblasts and intracellular and extracellular TAGE, which are secreted from certain organs, remain unclear. We examined the cytotoxicity of intracellular TAGE by a slot blot analysis, and TAGE-modified bovine serum albumin (TAGE-BSA), a model of extracellular TAGE, in normal human cardiac fibroblasts (HCF). Intracellular TAGE induced cell death in normal HCF, whereas TAGE-BSA did not, even at aberrantly high non-physiological levels. Therefore, only intracellular TAGE induced cell death in HCF under physiological conditions, possibly inhibiting the role of HCF.
Collapse
Affiliation(s)
- Takanobu Takata
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan; (A.S.-S.); (M.T.)
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan
- Correspondence: ; Tel.: +81-76-2211
| | - Akiko Sakasai-Sakai
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan; (A.S.-S.); (M.T.)
| | - Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan; (A.S.-S.); (M.T.)
| |
Collapse
|
6
|
Ooi H, Nasu R, Furukawa A, Takeuchi M, Koriyama Y. Pyridoxamine and Aminoguanidine Attenuate the Abnormal Aggregation of β-Tubulin and Suppression of Neurite Outgrowth by Glyceraldehyde-Derived Toxic Advanced Glycation End-Products. Front Pharmacol 2022; 13:921611. [PMID: 35721214 PMCID: PMC9204210 DOI: 10.3389/fphar.2022.921611] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/13/2022] [Indexed: 01/03/2023] Open
Abstract
Diabetes mellitus (DM) has been identified as a risk factor for the onset and progression of Alzheimer’s disease (AD). In our previous study, we demonstrated that glyceraldehyde (GA)-derived toxic advanced glycation end-products (toxic AGEs, TAGE) induced similar alterations to those observed in AD. GA induced dysfunctional neurite outgrowth via TAGE-β-tubulin aggregation, which resulted in the TAGE-dependent abnormal aggregation of β-tubulin and tau phosphorylation in human neuroblastoma SH-SY5Y cells. However, the effects of inhibitors of AGE formation on dysfunctional neurite outgrowth caused by GA-induced abnormalities in the aggregation of β-tubulin and tau phosphorylation remain unknown. Aminoguanidine (AG), an AGE inhibitor, and pyridoxamine (PM), a natural form of vitamin B6 (VB6), are effective AGE inhibitors. Therefore, the present study investigated whether AG or PM ameliorate TAGE-β-tubulin aggregation and the suppression of neurite outgrowth by GA. The results obtained showed that AG and PM inhibited the formation of TAGE-β-tubulin, mitigated the GA-induced suppression of neurite outgrowth, and reduced GA-mediated increases in tau phosphorylation levels. Collectively, these results suggest the potential of AG and PM to prevent the DM-associated onset and progression of AD.
Collapse
Affiliation(s)
- Hayahide Ooi
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
| | - Ryuto Nasu
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
| | - Ayako Furukawa
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
| | - Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Japan
| | - Yoshiki Koriyama
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Japan
- *Correspondence: Yoshiki Koriyama,
| |
Collapse
|
7
|
Takeuchi M, Sakasai-Sakai A, Takata T, Takino JI, Koriyama Y, Kikuchi C, Furukawa A, Nagamine K, Hori T, Matsunaga T. Intracellular Toxic AGEs (TAGE) Triggers Numerous Types of Cell Damage. Biomolecules 2021; 11:biom11030387. [PMID: 33808036 PMCID: PMC8001776 DOI: 10.3390/biom11030387] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
The habitual intake of large amounts of sugar, which has been implicated in the onset/progression of lifestyle-related diseases (LSRD), induces the excessive production of glyceraldehyde (GA), an intermediate of sugar metabolism, in neuronal cells, hepatocytes, and cardiomyocytes. Reactions between GA and intracellular proteins produce toxic advanced glycation end-products (toxic AGEs, TAGE), the accumulation of which contributes to various diseases, such as Alzheimer’s disease, non-alcoholic steatohepatitis, and cardiovascular disease. The cellular leakage of TAGE affects the surrounding cells via the receptor for AGEs (RAGE), thereby promoting the onset/progression of LSRD. We demonstrated that the intracellular accumulation of TAGE triggered numerous cellular disorders, and also that TAGE leaked into the extracellular space, thereby increasing extracellular TAGE levels in circulating fluids. Intracellular signaling and the production of reactive oxygen species are affected by extracellular TAGE and RAGE interactions, which, in turn, facilitate the intracellular generation of TAGE, all of which may contribute to the pathological changes observed in LSRD. In this review, we discuss the relationships between intracellular TAGE levels and numerous types of cell damage. The novel concept of the “TAGE theory” is expected to open new perspectives for research into LSRD.
Collapse
Affiliation(s)
- Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan; (A.S.-S.); (T.T.)
- Correspondence: ; Tel.: +81-76-218-8456
| | - Akiko Sakasai-Sakai
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan; (A.S.-S.); (T.T.)
| | - Takanobu Takata
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada-machi, Ishikawa 920-0293, Japan; (A.S.-S.); (T.T.)
| | - Jun-ichi Takino
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (J.-i.T.); (T.H.)
| | - Yoshiki Koriyama
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8670, Japan; (Y.K.); (A.F.)
| | - Chigusa Kikuchi
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (C.K.); (T.M.)
| | - Ayako Furukawa
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka, Mie 513-8670, Japan; (Y.K.); (A.F.)
| | - Kentaro Nagamine
- Department of Clinical Nutrition, Faculty of Health Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan;
| | - Takamitsu Hori
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (J.-i.T.); (T.H.)
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (C.K.); (T.M.)
| |
Collapse
|
8
|
Crivelli JJ, Mitchell T, Knight J, Wood KD, Assimos DG, Holmes RP, Fargue S. Contribution of Dietary Oxalate and Oxalate Precursors to Urinary Oxalate Excretion. Nutrients 2020; 13:nu13010062. [PMID: 33379176 PMCID: PMC7823532 DOI: 10.3390/nu13010062] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/23/2020] [Accepted: 12/25/2020] [Indexed: 02/07/2023] Open
Abstract
Kidney stone disease is increasing in prevalence, and the most common stone composition is calcium oxalate. Dietary oxalate intake and endogenous production of oxalate are important in the pathophysiology of calcium oxalate stone disease. The impact of dietary oxalate intake on urinary oxalate excretion and kidney stone disease risk has been assessed through large cohort studies as well as smaller studies with dietary control. Net gastrointestinal oxalate absorption influences urinary oxalate excretion. Oxalate-degrading bacteria in the gut microbiome, especially Oxalobacter formigenes, may mitigate stone risk through reducing net oxalate absorption. Ascorbic acid (vitamin C) is the main dietary precursor for endogenous production of oxalate with several other compounds playing a lesser role. Renal handling of oxalate and, potentially, renal synthesis of oxalate may contribute to stone formation. In this review, we discuss dietary oxalate and precursors of oxalate, their pertinent physiology in humans, and what is known about their role in kidney stone disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Ross P. Holmes
- Correspondence: ; Tel.: +1-(205)-996-8765; Fax: +1-(205)-934-4933
| | | |
Collapse
|
9
|
Nasu R, Furukawa A, Suzuki K, Takeuchi M, Koriyama Y. The Effect of Glyceraldehyde-Derived Advanced Glycation End Products on β-Tubulin-Inhibited Neurite Outgrowth in SH-SY5Y Human Neuroblastoma Cells. Nutrients 2020; 12:nu12102958. [PMID: 32992566 PMCID: PMC7601248 DOI: 10.3390/nu12102958] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Nutritional factors can affect the risk of developing neurological disorders and their rate of progression. In particular, abnormalities of carbohydrate metabolism in diabetes mellitus patients lead to an increased risk of neurological disorders such as Alzheimer’s disease (AD). In this study, we investigated the relationship between nervous system disorder and the pathogenesis of AD by exposing SH-SY5Y neuroblastoma cells to glyceraldehyde (GA). We previously reported that GA-derived toxic advanced glycation end products (toxic AGEs, TAGE) induce AD-like alterations including intracellular tau phosphorylation. However, the role of TAGE and their target molecules in the pathogenesis of AD remains unclear. In this study, we investigated the target protein for TAGE by performing two-dimensional immunoblot analysis with anti-TAGE antibody and mass spectrometry and identified β-tubulin as one of the targets. GA treatment induced TAGE-β-tubulin formation and abnormal aggregation of β-tubulin, and inhibited neurite outgrowth in SH-SY5Y cells. On the other hand, glucose-derived AGEs were also involved in developing AD. However, glucose did not make abnormal aggregation of β-tubulin and did not inhibit neurite outgrowth. Understanding the underlying mechanism of TAGE-β-tubulin formation by GA and its role in neurodegeneration may aid in the development of novel therapeutics and neuroprotection strategies.
Collapse
Affiliation(s)
- Ryuto Nasu
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka 513-8670, Japan; (R.N.); (A.F.); (K.S.)
| | - Ayako Furukawa
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka 513-8670, Japan; (R.N.); (A.F.); (K.S.)
| | - Keita Suzuki
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka 513-8670, Japan; (R.N.); (A.F.); (K.S.)
| | - Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa 920-0293, Japan;
| | - Yoshiki Koriyama
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka 513-8670, Japan; (R.N.); (A.F.); (K.S.)
- Correspondence:
| |
Collapse
|
10
|
Takayasu S, Kamba A, Yoshida K, Terui K, Watanuki Y, Ishigame N, Mizushiri S, Tomita T, Nakamura K, Yasui-Furukori N, Daimon M. Secondary oxalosis induced by xylitol concurrent with lithium-induced nephrogenic diabetes insipidus: a case report. BMC Nephrol 2020; 21:157. [PMID: 32357847 PMCID: PMC7195762 DOI: 10.1186/s12882-020-01814-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 04/16/2020] [Indexed: 12/04/2022] Open
Abstract
Background Xylitol is an approved food additive that is widely used as a sweetener in many manufactured products. It is also used in pharmaceuticals. Secondary oxalosis resulting from high dietary oxalate has been reported. However, reported cases of oxalosis following xylitol infusion are rare. Case presentation A 39-year-old man with a 16-year history of organic psychiatric disorder was hospitalized for a laparoscopic cholecystectomy because of cholecystolithiasis. He had been treated with several antipsychotics and mood stabilizers, including lithium. The patient had polyuria (> 4000 mL/day) and his serum sodium levels ranged from 150 to 160 mmol/L. Urine osmolality was 141 mOsm/L, while serum arginine vasopressin level was 6.4 pg/mL. The patient was diagnosed with nephrogenic diabetes insipidus (NDI), and lithium was gradually discontinued. Postoperative urine volumes increased further to a maximum of 10,000 mL/day, and up to 10,000 mL/day of 5% xylitol was administered. The patient’s consciousness level declined and serum creatinine increased to 4.74 mg/dL. This was followed by coma and metabolic acidosis. After continuous venous hemodiafiltration, serum sodium improved to the upper 140 mmol/L range and serum creatinine decreased to 1.25 mg/dL at discharge. However, polyuria and polydipsia of approximately 4000 mL/day persisted. Renal biopsy showed oxalate crystals and decreased expression of aquaporin-2 (AQP2) in the renal tubules. Urinary AQP2 was undetected. The patient was discharged on day 82 after admission. Conclusions Our patient was diagnosed with lithium-induced NDI and secondary oxalosis induced by excess xylitol infusion. NDI became apparent perioperatively because of fasting, and an overdose of xylitol infusion led to cerebrorenal oxalosis. Our patient received a maximum xylitol dose of 500 g/day and a total dose of 2925 g. Patients receiving lithium therapy must be closely monitored during the perioperative period, and rehydration therapy using xylitol infusion should be avoided in such cases.
Collapse
Affiliation(s)
- Shinobu Takayasu
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan.
| | - Aya Kamba
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Kazutaka Yoshida
- Department of Neuropsychiatry, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Ken Terui
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Yutaka Watanuki
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Noriko Ishigame
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Satoru Mizushiri
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Tetsu Tomita
- Department of Neuropsychiatry, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Kazuhiko Nakamura
- Department of Neuropsychiatry, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Norio Yasui-Furukori
- Department of Neuropsychiatry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi, 321-0293, Japan
| | - Makoto Daimon
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine and Hospital, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| |
Collapse
|
11
|
Takata T, Sakasai-Sakai A, Takeuchi M. Impact of intracellular toxic advanced glycation end-products (TAGE) on murine myoblast cell death. Diabetol Metab Syndr 2020; 12:54. [PMID: 32684984 PMCID: PMC7362572 DOI: 10.1186/s13098-020-00561-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Sarcopenia is a progressive condition that is characterized by decreases in skeletal muscle mass and function. Although sarcopenia is associated with lifestyle-related diseases (LSRD), the mechanisms underlying cell death in myoblasts, which differentiate to myotubes, remain unclear. We previously designated glyceraldehyde (an intermediate of glucose/fructose metabolism)-derived advanced glycation end-products (AGEs) as toxic AGEs (TAGE) because of their cytotoxicity and involvement in LSRD, and hypothesized that TAGE contribute to cell death in myoblasts. METHODS C2C12 cells, which are murine myoblasts, were treated with 0, 0.5, 1, 1.5, and 2 mM glyceraldehyde for 24 h. Cell viability and intracellular TAGE were then assessed using 5-[2,4,-bis(sodioxysulfonyl)phenyl]-3-(2-methoxy-4-nitrophenyl)-2-(4-nitrophenyl)-2H-tetrazole-3-ium (WST-8) and slot blot assays. Cells were pretreated with 8 mM aminoguanidine, an inhibitor of AGE production, for 2 h, followed by 0, 1.5, and 2 mM glyceraldehyde for 24 h. Cell viability and intracellular TAGE levels were then assessed. Serum TAGE levels in STAM mice, in which there were four stages (no steatosis, simple steatosis, steatohepatitis, and fibrosis), were measured using a competitive enzyme-linked immunosorbent assay. Results were expressed as TAGE units (U) per milliliter of serum, with 1 U corresponding to 1.0 μg of glyceraldehyde-derived AGE-bovine serum albumin (BSA) (TAGE-BSA). The viability of cells treated with 20, 50, and 100 μg/mL non-glycated BSA and TAGE-BSA for 24 h was assessed using the WST-8 assay. RESULTS In C2C12 cells treated with 1.5 and 2 mM glyceraldehyde, cell viability decreased to 47.7% (p = 0.0021) and 5.0% (p = 0.0001) and intracellular TAGE levels increased to 6.0 and 15.9 μg/mg protein, respectively. Changes in cell viability and TAGE production were completely inhibited by 8 mM aminoguanidine. Serum TAGE levels at the steatohepatitis and fibrosis stages were 10.51 ± 1.16 and 10.44 ± 0.95 U/mL, respectively, and were higher than those at the no steatosis stage (7.27 ± 0.18 U/mL). Cell death was not induced by 20 or 50 μg/mL TAGE-BSA. The viabilities of C2C12 cells treated with 100 μg/mL non-glycated BSA and TAGE-BSA were 105.0% (p = 0.2890) and 85.3% (p = 0.0217), respectively. CONCLUSION Intracellular TAGE strongly induced cell death in C2C12 cells and may also induce myoblast cell death in LSRD model mice.
Collapse
Affiliation(s)
- Takanobu Takata
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa, 920-0293 Japan
| | - Akiko Sakasai-Sakai
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa, 920-0293 Japan
| | - Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa, 920-0293 Japan
| |
Collapse
|
12
|
Uranukul B, Woolston BM, Fink GR, Stephanopoulos G. Biosynthesis of monoethylene glycol in Saccharomyces cerevisiae utilizing native glycolytic enzymes. Metab Eng 2018; 51:20-31. [PMID: 30268818 DOI: 10.1016/j.ymben.2018.09.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/07/2018] [Accepted: 09/25/2018] [Indexed: 10/28/2022]
Abstract
Monoethylene glycol (MEG) is an important commodity chemical with applications in numerous industrial processes, primarily in the manufacture of polyethylene terephthalate (PET) polyester used in packaging applications. In the drive towards a sustainable chemical industry, bio-based production of MEG from renewable biomass has attracted growing interest. Recent attempts for bio-based MEG production have investigated metabolic network modifications in Escherichia coli, specifically rewiring the xylose assimilation pathways for the synthesis of MEG. In the present study, we examined the suitability of Saccharomyces cerevisiae, a preferred organism for industrial applications, as platform for MEG biosynthesis. Based on combined genetic, biochemical and fermentation studies, we report evidence for the existence of an endogenous biosynthetic route for MEG production from D-xylose in S. cerevisiae which consists of phosphofructokinase and fructose-bisphosphate aldolase, the two key enzymes in the glycolytic pathway. Further metabolic engineering and process optimization yielded a strain capable of producing up to 4.0 g/L MEG, which is the highest titer reported in yeast to-date.
Collapse
Affiliation(s)
- Boonsom Uranukul
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Whitehead Institute for Biomedical Research, Cambridge, MA 02139, United States
| | - Benjamin M Woolston
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Gerald R Fink
- Whitehead Institute for Biomedical Research, Cambridge, MA 02139, United States
| | - Gregory Stephanopoulos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States.
| |
Collapse
|
13
|
Tappy L. Fructose metabolism and noncommunicable diseases: recent findings and new research perspectives. Curr Opin Clin Nutr Metab Care 2018; 21:214-222. [PMID: 29406418 DOI: 10.1097/mco.0000000000000460] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW There is increasing concern that dietary fructose may contribute to the development of noncommunicable diseases. This review identifies major new findings related to fructose's physiological or adverse effects. RECENT FINDINGS Fructose is mainly processed in splanchnic organs (gut, liver, kidneys) to glucose, lactate, and fatty acids, which can then be oxidized in extrasplanchnic organs and tissues. There is growing evidence that splanchnic lactate production, linked to extrasplanchnic lactate metabolism, represents a major fructose disposal pathway during and after exercise. Chronic excess fructose intake can be directly responsible for an increase in intrahepatic fat concentration and for the development of hepatic, but not muscle insulin resistance. Although it has long been thought that fructose was exclusively metabolized in splanchnic organs, several recent reports provide indirect that some fructose may also be metabolized in extrasplanchnic cells, such as adipocytes, muscle, or brain cells; the quantity of fructose directly metabolized in extrasplanchnic cells, and its physiological consequences, remain however unknown. There is also growing evidence that endogenous fructose production from glucose occurs in humans and may have important physiological functions, but may also be associated with adverse health effects. SUMMARY Fructose is a physiological nutrient which, when consumed in excess, may have adverse metabolic effects, mainly in the liver (hepatic insulin resistance and fat storage). There is also concern that exogenous or endogenously produced fructose may be directly metabolized in extrasplanchnic cells in which it may exert adverse metabolic effects.
Collapse
Affiliation(s)
- Luc Tappy
- Physiology Department, Faculty of Biology and Medicine, University of Lausanne, Lausanne
- Metabolic Center, Hôpital Intercantonal de la Broye, Estavayer-le-lac, Switzerland
| |
Collapse
|
14
|
Deletion of transketolase triggers a stringent metabolic response in promastigotes and loss of virulence in amastigotes of Leishmania mexicana. PLoS Pathog 2018; 14:e1006953. [PMID: 29554142 PMCID: PMC5882173 DOI: 10.1371/journal.ppat.1006953] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/03/2018] [Accepted: 02/28/2018] [Indexed: 11/22/2022] Open
Abstract
Transketolase (TKT) is part of the non-oxidative branch of the pentose phosphate pathway (PPP). Here we describe the impact of removing this enzyme from the pathogenic protozoan Leishmania mexicana. Whereas the deletion had no obvious effect on cultured promastigote forms of the parasite, the Δtkt cells were not virulent in mice. Δtkt promastigotes were more susceptible to oxidative stress and various leishmanicidal drugs than wild-type, and metabolomics analysis revealed profound changes to metabolism in these cells. In addition to changes consistent with those directly related to the role of TKT in the PPP, central carbon metabolism was substantially decreased, the cells consumed significantly less glucose, flux through glycolysis diminished, and production of the main end products of metabolism was decreased. Only minor changes in RNA abundance from genes encoding enzymes in central carbon metabolism, however, were detected although fructose-1,6-bisphosphate aldolase activity was decreased two-fold in the knock-out cell line. We also showed that the dual localisation of TKT between cytosol and glycosomes is determined by the C-terminus of the enzyme and by engineering different variants of the enzyme we could alter its sub-cellular localisation. However, no effect on the overall flux of glucose was noted irrespective of whether the enzyme was found uniquely in either compartment, or in both. Leishmania parasites endanger over 1 billion people worldwide, infecting 300,000 people and causing 20,000 deaths annually. In this study, we scrutinized metabolism in Leishmania mexicana after deletion of the gene encoding transketolase (TKT), an enzyme involved in sugar metabolism via the pentose phosphate pathway which plays key roles in creating ribose 5-phosphate for nucleotide synthesis and also defence against oxidative stress. The insect stage of the parasite, grown in culture medium, did not suffer from any obvious growth defect after the gene was deleted. However, its metabolism changed dramatically, with metabolomics indicating profound changes to flux through the pentose phosphate pathway: decreased glucose consumption, and generally enhanced efficiency in using metabolic substrates with reduced secretion of partially oxidised end products of metabolism. This ‘stringent’ metabolism is reminiscent of the mammalian stage parasites. The cells were also more sensitive to oxidative stress inducing agents and leishmanicidal drugs. Crucially, mice inoculated with the TKT knock-out parasites did not develop an infection pointing to the enzyme playing a key role in allowing the parasites to remain viable in the host, indicating that TKT may be considered a useful target for development of new drugs against leishmaniasis.
Collapse
|
15
|
Huard K, Ahn K, Amor P, Beebe DA, Borzilleri KA, Chrunyk BA, Coffey SB, Cong Y, Conn EL, Culp JS, Dowling MS, Gorgoglione MF, Gutierrez JA, Knafels JD, Lachapelle EA, Pandit J, Parris KD, Perez S, Pfefferkorn JA, Price DA, Raymer B, Ross TT, Shavnya A, Smith AC, Subashi TA, Tesz GJ, Thuma BA, Tu M, Weaver JD, Weng Y, Withka JM, Xing G, Magee TV. Discovery of Fragment-Derived Small Molecules for in Vivo Inhibition of Ketohexokinase (KHK). J Med Chem 2017; 60:7835-7849. [PMID: 28853885 DOI: 10.1021/acs.jmedchem.7b00947] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Increased fructose consumption and its subsequent metabolism have been implicated in hepatic steatosis, dyslipidemia, obesity, and insulin resistance in humans. Since ketohexokinase (KHK) is the principal enzyme responsible for fructose metabolism, identification of a selective KHK inhibitor may help to further elucidate the effect of KHK inhibition on these metabolic disorders. Until now, studies on KHK inhibition with small molecules have been limited due to the lack of viable in vivo pharmacological tools. Herein we report the discovery of 12, a selective KHK inhibitor with potency and properties suitable for evaluating KHK inhibition in rat models. Key structural features interacting with KHK were discovered through fragment-based screening and subsequent optimization using structure-based drug design, and parallel medicinal chemistry led to the identification of pyridine 12.
Collapse
Affiliation(s)
- Kim Huard
- Medicine Design, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Kay Ahn
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Paul Amor
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - David A Beebe
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Kris A Borzilleri
- Structural Biology and Biophysics, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Boris A Chrunyk
- Structural Biology and Biophysics, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Steven B Coffey
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yang Cong
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Edward L Conn
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jeffrey S Culp
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew S Dowling
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew F Gorgoglione
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jemy A Gutierrez
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - John D Knafels
- Structural Biology and Biophysics, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Erik A Lachapelle
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jayvardhan Pandit
- Structural Biology and Biophysics, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kevin D Parris
- Structural Biology and Biophysics, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sylvie Perez
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jeffrey A Pfefferkorn
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - David A Price
- Medicine Design, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Brian Raymer
- Medicine Design, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Trenton T Ross
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Andre Shavnya
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Aaron C Smith
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Timothy A Subashi
- Structural Biology and Biophysics, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gregory J Tesz
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Benjamin A Thuma
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Meihua Tu
- Medicine Design, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - John D Weaver
- Medicine Design, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Yan Weng
- Medicine Design, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jane M Withka
- Structural Biology and Biophysics, Pfizer Inc. , Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gang Xing
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Thomas V Magee
- Internal Medicine, Pfizer Inc. , 1 Portland Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
16
|
Cam Y, Alkim C, Trichez D, Trebosc V, Vax A, Bartolo F, Besse P, François JM, Walther T. Engineering of a Synthetic Metabolic Pathway for the Assimilation of (d)-Xylose into Value-Added Chemicals. ACS Synth Biol 2016; 5:607-18. [PMID: 26186096 DOI: 10.1021/acssynbio.5b00103] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A synthetic pathway for (d)-xylose assimilation was stoichiometrically evaluated and implemented in Escherichia coli strains. The pathway proceeds via isomerization of (d)-xylose to (d)-xylulose, phosphorylation of (d)-xylulose to obtain (d)-xylulose-1-phosphate (X1P), and aldolytic cleavage of the latter to yield glycolaldehyde and DHAP. Stoichiometric analyses showed that this pathway provides access to ethylene glycol with a theoretical molar yield of 1. Alternatively, both glycolaldehyde and DHAP can be converted to glycolic acid with a theoretical yield that is 20% higher than for the exclusive production of this acid via the glyoxylate shunt. Simultaneous expression of xylulose-1 kinase and X1P aldolase activities, provided by human ketohexokinase-C and human aldolase-B, respectively, restored growth of a (d)-xylulose-5-kinase mutant on xylose. This strain produced ethylene glycol as the major metabolic endproduct. Metabolic engineering provided strains that assimilated the entire C2 fraction into the central metabolism or that produced 4.3 g/L glycolic acid at a molar yield of 0.9 in shake flasks.
Collapse
Affiliation(s)
- Yvan Cam
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés (LISBP),, 31077 Toulouse, France
- UMR5504, CNRS, 31077 Toulouse, France
- TWB, 3 rue des Satellites, Canal Biotech Building 2, 31400 Toulouse, France
| | - Ceren Alkim
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés (LISBP),, 31077 Toulouse, France
- UMR5504, CNRS, 31077 Toulouse, France
- TWB, 3 rue des Satellites, Canal Biotech Building 2, 31400 Toulouse, France
| | - Debora Trichez
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés (LISBP),, 31077 Toulouse, France
- UMR5504, CNRS, 31077 Toulouse, France
- TWB, 3 rue des Satellites, Canal Biotech Building 2, 31400 Toulouse, France
| | - Vincent Trebosc
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés (LISBP),, 31077 Toulouse, France
- UMR5504, CNRS, 31077 Toulouse, France
- TWB, 3 rue des Satellites, Canal Biotech Building 2, 31400 Toulouse, France
| | - Amélie Vax
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés (LISBP),, 31077 Toulouse, France
- UMR5504, CNRS, 31077 Toulouse, France
- TWB, 3 rue des Satellites, Canal Biotech Building 2, 31400 Toulouse, France
| | - François Bartolo
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- Département Génie Mathématiques et Modélisation (GMM), 135 Avenue de Rangueil, 31077 Toulouse, France
| | - Philippe Besse
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- Département Génie Mathématiques et Modélisation (GMM), 135 Avenue de Rangueil, 31077 Toulouse, France
| | - Jean Marie François
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés (LISBP),, 31077 Toulouse, France
- UMR5504, CNRS, 31077 Toulouse, France
- TWB, 3 rue des Satellites, Canal Biotech Building 2, 31400 Toulouse, France
| | - Thomas Walther
- INSA, UPS, INP, LISBP, Université de Toulouse, 135 Avenue de Rangueil, 31077 Toulouse, France
- INRA, UMR792 Ingénierie des Systèmes Biologiques et des Procédés (LISBP),, 31077 Toulouse, France
- UMR5504, CNRS, 31077 Toulouse, France
- TWB, 3 rue des Satellites, Canal Biotech Building 2, 31400 Toulouse, France
| |
Collapse
|
17
|
Chomvong K, Bauer S, Benjamin DI, Li X, Nomura DK, Cate JHD. Bypassing the Pentose Phosphate Pathway: Towards Modular Utilization of Xylose. PLoS One 2016; 11:e0158111. [PMID: 27336308 PMCID: PMC4918971 DOI: 10.1371/journal.pone.0158111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/12/2016] [Indexed: 12/22/2022] Open
Abstract
The efficient use of hemicellulose in the plant cell wall is critical for the economic conversion of plant biomass to renewable fuels and chemicals. Previously, the yeast Saccharomyces cerevisiae has been engineered to convert the hemicellulose-derived pentose sugars xylose and arabinose to d-xylulose-5-phosphate for conversion via the pentose phosphate pathway (PPP). However, efficient pentose utilization requires PPP optimization and may interfere with its roles in NADPH and pentose production. Here, we developed an alternative xylose utilization pathway that largely bypasses the PPP. In the new pathway, d-xylulose is converted to d-xylulose-1-phosphate, a novel metabolite to S. cerevisiae, which is then cleaved to glycolaldehyde and dihydroxyacetone phosphate. This synthetic pathway served as a platform for the biosynthesis of ethanol and ethylene glycol. The use of d-xylulose-1-phosphate as an entry point for xylose metabolism opens the way for optimizing chemical conversion of pentose sugars in S. cerevisiae in a modular fashion.
Collapse
Affiliation(s)
- Kulika Chomvong
- Department of Plant and Microbial Biology, University of California, Berkeley, CA, United States of America
| | - Stefan Bauer
- Energy Biosciences Institute, Berkeley, CA, United States of America
| | - Daniel I. Benjamin
- Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, United States of America
| | - Xin Li
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, United States of America
| | - Daniel K. Nomura
- Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, United States of America
| | - Jamie H. D. Cate
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, United States of America
- Department of Chemistry, University of California, Berkeley, CA, United States of America
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States of America
- * E-mail:
| |
Collapse
|
18
|
Affiliation(s)
- Liuqing Wen
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Kenneth Huang
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Yunpeng Liu
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
- National
Glycoengineering Research Center, Shandong University, Jinan 250100, China
| | - Peng George Wang
- Department
of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
- National
Glycoengineering Research Center, Shandong University, Jinan 250100, China
| |
Collapse
|
19
|
Koriyama Y, Furukawa A, Muramatsu M, Takino JI, Takeuchi M. Glyceraldehyde caused Alzheimer's disease-like alterations in diagnostic marker levels in SH-SY5Y human neuroblastoma cells. Sci Rep 2015; 5:13313. [PMID: 26304819 PMCID: PMC4548441 DOI: 10.1038/srep13313] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/21/2015] [Indexed: 01/12/2023] Open
Abstract
Clinical evidence has implicated diabetes mellitus as one of the risk factors for the development and progression of Alzheimer’s disease (AD). However, the neurotoxic pathway activated due to abnormalities in glucose metabolism has not yet been identified in AD. In order to investigate the relationship between impaired cerebral glucose metabolism and the pathophysiology of AD, SH-SY5Y human neuroblastoma cells were exposed to glyceraldehyde (GA), an inhibitor of glycolysis. GA induced the production of GA-derived advanced glycation end-products (GA-AGEs) and cell apoptosis, glycolytic inhibition, decreases in the medium concentrations of diagnostic markers of AD, such as amyloid β 1-42 (Aβ42), and increases in tau phosphorylation. These results suggest that the production of GA-AGEs and/or inhibition of glycolysis induce AD-like alterations, and this model may be useful for examining the pathophysiology of AD.
Collapse
Affiliation(s)
- Yoshiki Koriyama
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, 513-8670, Japan
| | - Ayako Furukawa
- Graduate School and Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, 513-8670, Japan
| | - Michiru Muramatsu
- Department of Pathophysiological Science, Faculty of Pharmaceutical Science, Hokuriku University, Kanazawa, Ishikawa, 920-1181, Japan
| | - Jun-ichi Takino
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hiroshima International University, Kure, Hiroshima, 737-0112, Japan
| | - Masayoshi Takeuchi
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada-machi, Ishikawa, 920-0293, Japan
| |
Collapse
|
20
|
Wen L, Huang K, Wei M, Meisner J, Liu Y, Garner K, Zang L, Wang X, Li X, Fang J, Zhang H, Wang PG. Facile Enzymatic Synthesis of Ketoses. Angew Chem Int Ed Engl 2015; 54:12654-8. [PMID: 26275233 DOI: 10.1002/anie.201505714] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Indexed: 11/12/2022]
Abstract
Studies of rare ketoses have been hampered by a lack of efficient preparation methods. A convenient, efficient, and cost-effective platform for the facile synthesis of ketoses is described. This method enables the preparation of difficult-to-access ketopentoses and ketohexoses from common and inexpensive starting materials with high yield and purity and without the need for a tedious isomer separation step.
Collapse
Affiliation(s)
- Liuqing Wen
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA)
| | - Kenneth Huang
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA)
| | - Mohui Wei
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA)
| | - Jeffrey Meisner
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA).,Current Address: Division of Pulmonary, Allergy and Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322 (USA)
| | - Yunpeng Liu
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA)
| | - Kristina Garner
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA)
| | - Lanlan Zang
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA)
| | - Xuan Wang
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA)
| | - Xu Li
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA)
| | - Junqiang Fang
- National Glycoengineering Research Center, Shandong University, Jinan 250100 (China)
| | - Houcheng Zhang
- National Glycoengineering Research Center, Shandong University, Jinan 250100 (China)
| | - Peng George Wang
- Department of Chemistry and Center for Therapeutics and Diagnostics, Georgia State University, Atlanta, GA 30303 (USA). .,National Glycoengineering Research Center, Shandong University, Jinan 250100 (China).
| |
Collapse
|
21
|
Wen L, Huang K, Wei M, Meisner J, Liu Y, Garner K, Zang L, Wang X, Li X, Fang J, Zhang H, Wang PG. Facile Enzymatic Synthesis of Ketoses. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201505714] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
22
|
Tan B, Dong S, Shepard RL, Kays L, Roth KD, Geeganage S, Kuo MS, Zhao G. Inhibition of Nicotinamide Phosphoribosyltransferase (NAMPT), an Enzyme Essential for NAD+ Biosynthesis, Leads to Altered Carbohydrate Metabolism in Cancer Cells. J Biol Chem 2015; 290:15812-15824. [PMID: 25944913 DOI: 10.1074/jbc.m114.632141] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Indexed: 12/18/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) has been extensively studied due to its essential role in NAD(+) biosynthesis in cancer cells and the prospect of developing novel therapeutics. To understand how NAMPT regulates cellular metabolism, we have shown that the treatment with FK866, a specific NAMPT inhibitor, leads to attenuation of glycolysis by blocking the glyceraldehyde 3-phosphate dehydrogenase step (Tan, B., Young, D. A., Lu, Z. H., Wang, T., Meier, T. I., Shepard, R. L., Roth, K., Zhai, Y., Huss, K., Kuo, M. S., Gillig, J., Parthasarathy, S., Burkholder, T. P., Smith, M. C., Geeganage, S., and Zhao, G. (2013) Pharmacological inhibition of nicotinamide phosphoribosyltransferase (NAMPT), an enzyme essential for NAD(+) biosynthesis, in human cancer cells: metabolic basis and potential clinical implications. J. Biol. Chem. 288, 3500-3511). Due to technical limitations, we failed to separate isotopomers of phosphorylated sugars. In this study, we developed an enabling LC-MS methodology. Using this, we confirmed the previous findings and also showed that NAMPT inhibition led to accumulation of fructose 1-phosphate and sedoheptulose 1-phosphate but not glucose 6-phosphate, fructose 6-phosphate, and sedoheptulose 7-phosphate as previously thought. To investigate the metabolic basis of the metabolite formation, we carried out biochemical and cellular studies and established the following. First, glucose-labeling studies indicated that fructose 1-phosphate was derived from dihydroxyacetone phosphate and glyceraldehyde, and sedoheptulose 1-phosphate was derived from dihydroxyacetone phosphate and erythrose via an aldolase reaction. Second, biochemical studies showed that aldolase indeed catalyzed these reactions. Third, glyceraldehyde- and erythrose-labeling studies showed increased incorporation of corresponding labels into fructose 1-phosphate and sedoheptulose 1-phosphate in FK866-treated cells. Fourth, NAMPT inhibition led to increased glyceraldehyde and erythrose levels in the cell. Finally, glucose-labeling studies showed accumulated fructose 1,6-bisphosphate in FK866-treated cells mainly derived from dihydroxyacetone phosphate and glyceraldehyde 3-phosphate. Taken together, this study shows that NAMPT inhibition leads to attenuation of glycolysis, resulting in further perturbation of carbohydrate metabolism in cancer cells. The potential clinical implications of these findings are also discussed.
Collapse
Affiliation(s)
- Bo Tan
- Discovery Chemistry, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Sucai Dong
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Robert L Shepard
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Lisa Kays
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Kenneth D Roth
- Discovery Chemistry, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Sandaruwan Geeganage
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285
| | - Ming-Shang Kuo
- Discovery Chemistry, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285.
| | - Genshi Zhao
- Cancer Research, Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285.
| |
Collapse
|
23
|
Hassel B, Elsais A, Frøland AS, Taubøll E, Gjerstad L, Quan Y, Dingledine R, Rise F. Uptake and metabolism of fructose by rat neocortical cells in vivo and by isolated nerve terminals in vitro. J Neurochem 2015; 133:572-81. [PMID: 25708447 DOI: 10.1111/jnc.13079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/26/2015] [Accepted: 02/17/2015] [Indexed: 12/11/2022]
Abstract
Fructose reacts spontaneously with proteins in the brain to form advanced glycation end products (AGE) that may elicit neuroinflammation and cause brain pathology, including Alzheimer's disease. We investigated whether fructose is eliminated by oxidative metabolism in neocortex. Injection of [(14) C]fructose or its AGE-prone metabolite [(14) C]glyceraldehyde into rat neocortex in vivo led to formation of (14) C-labeled alanine, glutamate, aspartate, GABA, and glutamine. In isolated neocortical nerve terminals, [(14) C]fructose-labeled glutamate, GABA, and aspartate, indicating uptake of fructose into nerve terminals and oxidative fructose metabolism in these structures. This was supported by high expression of hexokinase 1, which channels fructose into glycolysis, and whose activity was similar with fructose or glucose as substrates. By contrast, the fructose-specific ketohexokinase was weakly expressed. The fructose transporter Glut5 was expressed at only 4% of the level of neuronal glucose transporter Glut3, suggesting transport across plasma membranes of brain cells as the limiting factor in removal of extracellular fructose. The genes encoding aldose reductase and sorbitol dehydrogenase, enzymes of the polyol pathway that forms glucose from fructose, were expressed in rat neocortex. These results point to fructose being transported into neocortical cells, including nerve terminals, and that it is metabolized and thereby detoxified primarily through hexokinase activity. We asked how the brain handles fructose, which may react spontaneously with proteins to form 'advanced glycation end products' and trigger inflammation. Neocortical cells took up and metabolized extracellular fructose oxidatively in vivo, and isolated nerve terminals did so in vitro. The low expression of fructose transporter Glut5 limited uptake of extracellular fructose. Hexokinase was a main pathway for fructose metabolism, but ketohexokinase (which leads to glyceraldehyde formation) was expressed too. Neocortical cells also took up and metabolized glyceraldehyde oxidatively.
Collapse
Affiliation(s)
- Bjørnar Hassel
- Department of Neurology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Norwegian Defence Research Establishment, Kjeller, Norway
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Fernández-Novell JM, Ramió-Lluch L, Orozco A, Gómez-Foix AM, Guinovart JJ, Rodríguez-Gil JE. Glucose and fructose have sugar-specific effects in both liver and skeletal muscle in vivo: a role for liver fructokinase. PLoS One 2014; 9:e109726. [PMID: 25330076 PMCID: PMC4201455 DOI: 10.1371/journal.pone.0109726] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/04/2014] [Indexed: 11/18/2022] Open
Abstract
We examined glucose and fructose effects on serine phosphorylation levels of a range of proteins in rat liver and muscle cells. For this, healthy adult rats were subjected to either oral glucose or fructose loads. A mini-array system was utilized to determine serine phosphorylation levels of liver and skeletal muscle proteins. A glucose oral load of 125 mg/100 g body weight (G 1/2) did not induce changes in phosphorylated serines of the proteins studied. Loading with 250 mg/100 g body weight of fructose (Fr), which induced similar glycemia levels as G 1/2, significantly increased serine phosphorylation of liver cyclin D3, PI3 kinase/p85, ERK-2, PTP2 and clusterin. The G 1/2 increased serine levels of the skeletal muscle proteins cyclin H, Cdk2, IRAK, total PKC, PTP1B, c-Raf 1, Ras and the β-subunit of the insulin receptor. The Fr induced a significant increase only in muscle serine phosphorylation of PI3 kinase/p85. The incubation of isolated rat hepatocytes with 10 mM glucose for 5 min significantly increased serine phosphorylation of 31 proteins. In contrast, incubation with 10 mM fructose produced less intense effects. Incubation with 10 mM glucose plus 75 µM fructose counteracted the effects of the incubation with glucose alone, except those on Raf-1 and Ras. Less marked effects were detected in cultured muscle cells incubated with 10 mM glucose or 10 mM glucose plus 75 µM fructose. Our results suggest that glucose and fructose act as specific functional modulators through a general mechanism that involves liver-generated signals, like micromolar fructosemia, which would inform peripheral tissues of the presence of either glucose- or fructose-derived metabolites.
Collapse
Affiliation(s)
| | - Laura Ramió-Lluch
- Dept. Animal Medicine and Surgery, Autonomous University of Barcelona, Bellaterra, Spain
| | - Anna Orozco
- Dept. Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Anna M. Gómez-Foix
- Dept. Biochemistry and Molecular Biology, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Joan J. Guinovart
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona Science Park, Barcelona, Spain
| | - Joan E. Rodríguez-Gil
- Dept. Animal Medicine and Surgery, Autonomous University of Barcelona, Bellaterra, Spain
- * E-mail:
| |
Collapse
|
25
|
Bunker RD, Bulloch EMM, Dickson JMJ, Loomes KM, Baker EN. Structure and function of human xylulokinase, an enzyme with important roles in carbohydrate metabolism. J Biol Chem 2012. [PMID: 23179721 DOI: 10.1074/jbc.m112.427997] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
D-Xylulokinase (XK; EC 2.7.1.17) catalyzes the ATP-dependent phosphorylation of d-xylulose (Xu) to produce xylulose 5-phosphate (Xu5P). In mammals, XK is the last enzyme in the glucuronate-xylulose pathway, active in the liver and kidneys, and is linked through its product Xu5P to the pentose-phosphate pathway. XK may play an important role in metabolic disease, given that Xu5P is a key regulator of glucose metabolism and lipogenesis. We have expressed the product of a putative human XK gene and identified it as the authentic human d-xylulokinase (hXK). NMR studies with a variety of sugars showed that hXK acts only on d-xylulose, and a coupled photometric assay established its key kinetic parameters as K(m)(Xu) = 24 ± 3 μm and k(cat) = 35 ± 5 s(-1). Crystal structures were determined for hXK, on its own and in complexes with Xu, ADP, and a fluorinated inhibitor. These reveal that hXK has a two-domain fold characteristic of the sugar kinase/hsp70/actin superfamily, with glycerol kinase as its closest relative. Xu binds to domain-I and ADP to domain-II, but in this open form of hXK they are 10 Å apart, implying that a large scale conformational change is required for catalysis. Xu binds in its linear keto-form, sandwiched between a Trp side chain and polar side chains that provide exquisite hydrogen bonding recognition. The hXK structure provides a basis for the design of specific inhibitors with which to probe its roles in sugar metabolism and metabolic disease.
Collapse
Affiliation(s)
- Richard D Bunker
- Maurice Wilkins Centre for Molecular Biodiscovery and School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | | | | | | | | |
Collapse
|
26
|
Mellor KM, Bell JR, Wendt IR, Davidoff AJ, Ritchie RH, Delbridge LMD. Fructose modulates cardiomyocyte excitation-contraction coupling and Ca²⁺ handling in vitro. PLoS One 2011; 6:e25204. [PMID: 21980397 PMCID: PMC3182977 DOI: 10.1371/journal.pone.0025204] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/30/2011] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND High dietary fructose has structural and metabolic cardiac impact, but the potential for fructose to exert direct myocardial action is uncertain. Cardiomyocyte functional responsiveness to fructose, and capacity to transport fructose has not been previously demonstrated. OBJECTIVE The aim of the present study was to seek evidence of fructose-induced modulation of cardiomyocyte excitation-contraction coupling in an acute, in vitro setting. METHODS AND RESULTS The functional effects of fructose on isolated adult rat cardiomyocyte contractility and Ca²⁺ handling were evaluated under physiological conditions (37°C, 2 mM Ca²⁺, HEPES buffer, 4 Hz stimulation) using video edge detection and microfluorimetry (Fura2) methods. Compared with control glucose (11 mM) superfusate, 2-deoxyglucose (2 DG, 11 mM) substitution prolonged both the contraction and relaxation phases of the twitch (by 16 and 36% respectively, p<0.05) and this effect was completely abrogated with fructose supplementation (11 mM). Similarly, fructose prevented the Ca²⁺ transient delay induced by exposure to 2 DG (time to peak Ca²⁺ transient: 2 DG: 29.0±2.1 ms vs. glucose: 23.6±1.1 ms vs. fructose +2 DG: 23.7±1.0 ms; p<0.05). The presence of the fructose transporter, GLUT5 (Slc2a5) was demonstrated in ventricular cardiomyocytes using real time RT-PCR and this was confirmed by conventional RT-PCR. CONCLUSION This is the first demonstration of an acute influence of fructose on cardiomyocyte excitation-contraction coupling. The findings indicate cardiomyocyte capacity to transport and functionally utilize exogenously supplied fructose. This study provides the impetus for future research directed towards characterizing myocardial fructose metabolism and understanding how long term high fructose intake may contribute to modulating cardiac function.
Collapse
Affiliation(s)
- Kimberley M Mellor
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
27
|
Diggle CP, Shires M, McRae C, Crellin D, Fisher J, Carr IM, Markham AF, Hayward BE, Asipu A, Bonthron DT. Both isoforms of ketohexokinase are dispensable for normal growth and development. Physiol Genomics 2010; 42A:235-43. [DOI: 10.1152/physiolgenomics.00128.2010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dietary fructose intake has dramatically increased over recent decades and is implicated in the high rates of obesity, hypertension, and type 2 diabetes (metabolic syndrome) in Western societies. The molecular determinants of this epidemiologic correlation are incompletely defined, but high-flux fructose catabolism initiated by ketohexokinase (Khk, fructokinase) is believed to be important. The Khk gene encodes two enzyme isoforms with distinctive substrate preferences, the independent physiological roles of which are unclear. To investigate this question, and for testing the importance of Khk in metabolic syndrome, isoform-selective genetic lesions would be valuable. Two deficiency alleles of the mouse Khk gene were designed. The first, Khk3a, uses targeted “knock-in” of a premature termination codon to induce a selective deficiency of the minor Khk-A isoform, preserving the major Khk-C isoform. The second, the KhkΔ allele, ablates both isoforms. Mice carrying each of these Khk-deficiency alleles were generated and validated at the DNA, RNA, and protein levels. Comparison between normal and knockout animals confirmed the specificity of the genetic lesions and allowed accurate analysis of the cellular distribution of Khk within tissues such as gut and liver. Both Khk3a/3a and KhkΔ/Δ homozygous mice were healthy and fertile and displayed minimal biochemical abnormalities under basal dietary conditions. These studies are the first demonstration that neither Khk isoform is required for normal growth and development. The new mouse models will allow direct testing of various hypotheses concerning the role of this enzyme in metabolic syndrome in humans and the value of Khk as a pharmacological target.
Collapse
Affiliation(s)
- C. P. Diggle
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, and
| | - M. Shires
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, and
| | - C. McRae
- School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - D. Crellin
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, and
| | - J. Fisher
- School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - I. M. Carr
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, and
| | - A. F. Markham
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, and
| | - B. E. Hayward
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, and
| | - A. Asipu
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, and
| | - D. T. Bonthron
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, and
| |
Collapse
|
28
|
Gibbs AC, Abad MC, Zhang X, Tounge BA, Lewandowski FA, Struble GT, Sun W, Sui Z, Kuo LC. Electron density guided fragment-based lead discovery of ketohexokinase inhibitors. J Med Chem 2010; 53:7979-91. [PMID: 21033679 DOI: 10.1021/jm100677s] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A fragment-based drug design paradigm has been successfully applied in the discovery of lead series of ketohexokinase inhibitors. The paradigm consists of three iterations of design, synthesis, and X-ray crystallographic screening to progress low molecular weight fragments to leadlike compounds. Applying electron density of fragments within the protein binding site as defined by X-ray crystallography, one can generate target specific leads without the use of affinity data. Our approach contrasts with most fragment-based drug design methodology where solution activity is a main design guide. Herein we describe the discovery of submicromolar ketohexokinase inhibitors with promising druglike properties.
Collapse
Affiliation(s)
- Alan C Gibbs
- Johnson & Johnson Pharmaceutical Research and Development, Welsh and McKean Roads, Spring House, Pennsylvania 19477, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Mellor KM, Ritchie RH, Davidoff AJ, Delbridge LMD. Elevated dietary sugar and the heart: experimental models and myocardial remodeling. Can J Physiol Pharmacol 2010; 88:525-40. [PMID: 20555422 DOI: 10.1139/y10-005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A dramatic rise in the prevalence of insulin resistance has been paralleled by increasing dietary consumption of sugar. The use of added sweeteners containing fructose (sucrose and high-fructose corn syrup) has increased by 25% over the past 3 decades. High fructose intake has the potential to adversely influence systemic and cellular metabolism via insulin resistance and glycolytic dysregulation. As a tissue that is both insulin sensitive and glycolysis dependent, the heart may be especially vulnerable to fructose over-consumption. In this review, experimental studies of elevated dietary sugar intake are evaluated, including sucrose and fructose dietary manipulation models. The possible role of the GLUT5 transporter as a mediator of cardiomyocyte fructose uptake is considered. The impact of dietary sucrose and fructose on cardiac insulin-dependent signaling in the context of perturbed systemic metabolic response is detailed. Myocardial dysfunction, modified growth, and oxidative stress responses associated with high dietary sugar intake are discussed. Finally, the involvement of the renin-angiotensin system in mediating fructose cardiopathology is considered. This review highlights the importance of obtaining new mechanistic data that can contribute to a more developed understanding of how high sugar intake directly contributes to structural and functional cardiomyopathy.
Collapse
Affiliation(s)
- Kimberley M Mellor
- Department of Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | | | | | | |
Collapse
|
30
|
Maier K, Hofmann U, Reuss M, Mauch K. Dynamics and control of the central carbon metabolism in hepatoma cells. BMC SYSTEMS BIOLOGY 2010; 4:54. [PMID: 20426867 PMCID: PMC2874527 DOI: 10.1186/1752-0509-4-54] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 04/28/2010] [Indexed: 02/08/2023]
Abstract
BACKGROUND The liver plays a major role in metabolism and performs a number of vital functions in the body. Therefore, the determination of hepatic metabolite dynamics and the analysis of the control of the respective biochemical pathways are of great pharmacological and medical importance. Extra- and intracellular time-series data from stimulus-response experiments are gaining in importance in the identification of in vivo metabolite dynamics, while dynamic network models are excellent tools for analyzing complex metabolic control patterns. This is the first study that has been undertaken on the data-driven identification of a dynamic liver central carbon metabolism model and its application in the analysis of the distribution of metabolic control in hepatoma cells. RESULTS Dynamic metabolite data were collected from HepG2 cells after they had been deprived of extracellular glucose. The concentration of 25 extra- and intracellular intermediates was quantified using HPLC, LC-MS-MS, and GC-MS. The in silico metabolite dynamics were in accordance with the experimental data. The central carbon metabolism of hepatomas was further analyzed with a particular focus on the control of metabolite concentrations and metabolic fluxes. It was observed that the enzyme glucose-6-phosphate dehydrogenase exerted substantial negative control over the glycolytic flux, whereas oxidative phosphorylation had a significant positive control. The control over the rate of NADPH consumption was found to be shared between the NADPH-demand itself (0.65) and the NADPH supply (0.38). CONCLUSIONS Based on time-series data, a dynamic central carbon metabolism model was developed for the investigation of new and complex metabolic control patterns in hepatoma cells. The control patterns found support the hypotheses that the glucose-6-phosphate dehydrogenase and the Warburg effect are promising targets for tumor treatment. The systems-oriented identification of metabolite dynamics is a first step towards the genome-based assessment of potential risks posed by nutrients and drugs.
Collapse
Affiliation(s)
- Klaus Maier
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Ute Hofmann
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Auerbachstrasse 112, 70376 Stuttgart, Germany
| | - Matthias Reuss
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Klaus Mauch
- Insilico Biotechnology AG, Nobelstrasse 15, 70569 Stuttgart, Germany
| |
Collapse
|
31
|
Keshari KR, Wilson DM, Chen AP, Bok R, Larson PEZ, Hu S, Van Criekinge M, Macdonald JM, Vigneron DB, Kurhanewicz J. Hyperpolarized [2-13C]-fructose: a hemiketal DNP substrate for in vivo metabolic imaging. J Am Chem Soc 2009; 131:17591-6. [PMID: 19860409 PMCID: PMC2796621 DOI: 10.1021/ja9049355] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hyperpolarized (13)C labeled molecular probes have been used to investigate metabolic pathways of interest as well as facilitate in vivo spectroscopic imaging by taking advantage of the dramatic signal enhancement provided by DNP. Due to the limited lifetime of the hyperpolarized nucleus, with signal decay dependent on T(1) relaxation, carboxylate carbons have been the primary targets for development of hyperpolarized metabolic probes. The use of these carbon nuclei makes it difficult to investigate upstream glycolytic processes, which have been related to both cancer metabolism as well as other metabolic abnormalities, such as fatty liver disease and diabetes. Glucose carbons have very short T(1)s (<1 s) and therefore cannot be used as an in vivo hyperpolarized metabolic probe of glycolysis. However, the pentose analogue fructose can also enter glycolysis through its phosphorylation by hexokinase and yield complementary information. The C(2) of fructose is a hemiketal that has a relatively longer relaxation time (approximately 16 s at 37 degrees C) and high solution state polarization (approximately 12%). Hyperpolarized [2-(13)C]-fructose was also injected into a transgenic model of prostate cancer (TRAMP) and demonstrated difference in uptake and metabolism in regions of tumor relative to surrounding tissue. Thus, this study demonstrates the first hyperpolarization of a carbohydrate carbon with a sufficient T(1) and solution state polarization for ex vivo spectroscopy and in vivo spectroscopic imaging studies.
Collapse
Affiliation(s)
- Kayvan R. Keshari
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 4 St., Byers Hall 203, San Francisco, California, United Sates 94158
- Department of Biomedical Engineering, University of North Carolina Chapel Hill (UNC), 152 MacNider Hall, Campus Box 7575, Chapel Hill, North Carolina, United States 27599
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 4 St., Byers Hall 203, San Francisco, California, United Sates 94158
| | - Albert P. Chen
- GE Healthcare, 333 Ravenswood Ave., Building 207, Menlo Park, CA 94025
| | - Robert Bok
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 4 St., Byers Hall 203, San Francisco, California, United Sates 94158
| | - Peder E. Z. Larson
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 4 St., Byers Hall 203, San Francisco, California, United Sates 94158
| | - Simon Hu
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 4 St., Byers Hall 203, San Francisco, California, United Sates 94158
| | - Mark Van Criekinge
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 4 St., Byers Hall 203, San Francisco, California, United Sates 94158
| | - Jeffrey M. Macdonald
- Department of Biomedical Engineering, University of North Carolina Chapel Hill (UNC), 152 MacNider Hall, Campus Box 7575, Chapel Hill, North Carolina, United States 27599
| | - Daniel B. Vigneron
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 4 St., Byers Hall 203, San Francisco, California, United Sates 94158
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco (UCSF), 1700 4 St., Byers Hall 203, San Francisco, California, United Sates 94158
| |
Collapse
|
32
|
Diggle CP, Shires M, Leitch D, Brooke D, Carr IM, Markham AF, Hayward BE, Asipu A, Bonthron DT. Ketohexokinase: expression and localization of the principal fructose-metabolizing enzyme. J Histochem Cytochem 2009; 57:763-74. [PMID: 19365088 DOI: 10.1369/jhc.2009.953190] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ketohexokinase (KHK, also known as fructokinase) initiates the pathway through which most dietary fructose is metabolized. Very little is known about the cellular localization of this enzyme. Alternatively spliced KHK-C and KHK-A mRNAs are known, but the existence of the KHK-A protein isoform has not been demonstrated in vivo. Using antibodies to KHK for immunohistochemistry and Western blotting of rodent tissues, including those from mouse knockouts, coupled with RT-PCR assays, we determined the distribution of the splice variants. The highly expressed KHK-C isoform localized to hepatocytes in the liver and to the straight segment of the proximal renal tubule. In both tissues, cytoplasmic and nuclear staining was observed. The KHK-A mRNA isoform was observed exclusively in a range of other tissues, and by Western blotting, the presence of endogenous immunoreactive KHK-A protein was shown for the first time, proving that the KHK-A mRNA is translated into KHK-A protein in vivo, and supporting the suggestion that this evolutionarily conserved isoform is physiologically functional. However, the low levels of KHK-A expression prevented its immunohistochemical localization within these tissues. Our results highlight that the use of in vivo biological controls (tissues from knockout animals) is required to distinguish genuine KHK immunoreactivity from experimental artifact.
Collapse
Affiliation(s)
- Christine P Diggle
- Leeds Institute of Molecular Medicine, University of Leeds, St James's University Hospital, Beckett Street, Leeds LS9 7TF, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Trinh CH, Asipu A, Bonthron DT, Phillips SEV. Structures of alternatively spliced isoforms of human ketohexokinase. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2009; 65:201-11. [PMID: 19237742 PMCID: PMC2651755 DOI: 10.1107/s0907444908041115] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Accepted: 12/05/2008] [Indexed: 11/10/2022]
Abstract
A molecular understanding of the unique aspects of dietary fructose metabolism may be the key to understanding and controlling the current epidemic of fructose-related obesity, diabetes and related adverse metabolic states in Western populations. Fructose catabolism is initiated by its phosphorylation to fructose 1-phosphate, which is performed by ketohexokinase (KHK). Here, the crystal structures of the two alternatively spliced isoforms of human ketohexokinase, hepatic KHK-C and the peripheral isoform KHK-A, and of the ternary complex of KHK-A with the substrate fructose and AMP-PNP are reported. The structure of the KHK-A ternary complex revealed an active site with both the substrate fructose and the ATP analogue in positions ready for phosphorylation following a reaction mechanism similar to that of the pfkB family of carbohydrate kinases. Hepatic KHK deficiency causes the benign disorder essential fructosuria. The effects of the disease-causing mutations (Gly40Arg and Ala43Thr) have been modelled in the context of the KHK structure.
Collapse
Affiliation(s)
- Chi H Trinh
- Astbury Centre for Structural Molecular Biology, Institute of Molecular and Cellular Biology, University of Leeds, Leeds, England
| | | | | | | |
Collapse
|
34
|
Cirillo P, Gersch MS, Mu W, Scherer PM, Kim KM, Gesualdo L, Henderson GN, Johnson RJ, Sautin YY. Ketohexokinase-dependent metabolism of fructose induces proinflammatory mediators in proximal tubular cells. J Am Soc Nephrol 2009; 20:545-53. [PMID: 19158351 DOI: 10.1681/asn.2008060576] [Citation(s) in RCA: 200] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Increased consumption of fructose may play an important role in the epidemic of metabolic syndrome and may presage the development of diabetes, cardiovascular disease, and chronic kidney disease. Once in the cell, fructose is phosphorylated by ketohexokinase (KHK), leading to consumption of ATP, formation of AMP, and generation of uric acid through xanthine oxidoreductase (XOR). This study aimed to examine the direct effects of fructose in human kidney proximal tubular cells (HK-2) and whether they are mediated by the fructose metabolism via KHK. At a similar concentration to that observed in peripheral blood after a meal, fructose induced production of monocyte chemotactic protein 1 (MCP-1) and reactive oxygen species in HK-2 cells. Knockdown of KHK by stable transfection with small hairpin RNA demonstrated that these processes were KHK dependent. Several antioxidants, including specific inhibitors of NADPH oxidase and XOR, prevented MCP-1 secretion. We detected XOR mRNA in HK-2 cells and confirmed its activity by identifying uric acid by mass spectrometry. Fructose increased intracellular uric acid, and uric acid induced production of MCP-1 as well. In summary, postprandial concentrations of fructose stimulate redox- and urate-dependent inflammatory mediators in proximal tubular cells.
Collapse
Affiliation(s)
- Pietro Cirillo
- Division of Nephrology, Hypertension and Transplantation, Department of Medicine, University of Florida, Gainesville, FL 32610-0224, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Faragó N, Kocsis GF, Fehér LZ, Csont T, Hackler L, Varga-Orvos Z, Csonka C, Kelemen JZ, Ferdinandy P, Puskás LG. Gene and protein expression changes in response to normoxic perfusion in mouse hearts. J Pharmacol Toxicol Methods 2008; 57:145-54. [DOI: 10.1016/j.vascn.2008.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 01/12/2008] [Indexed: 12/11/2022]
|
36
|
Pillai JB, Chen M, Rajamohan SB, Samant S, Pillai VB, Gupta M, Gupta MP. Activation of SIRT1, a class III histone deacetylase, contributes to fructose feeding-mediated induction of the alpha-myosin heavy chain expression. Am J Physiol Heart Circ Physiol 2008; 294:H1388-97. [PMID: 18192211 DOI: 10.1152/ajpheart.01339.2007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Fructose feeding has been shown to induce the cardiac alpha-myosin heavy chain (MHC) expression and protect the heart from ischemia- and reperfusion-mediated cell injury. This study was designed to investigate the mechanism involved in the effect of this sugar on MHC gene expression and cardiac protection. Adult mice were fed with a 6-propyl-2-thiouracil (PTU) diet or PTU combined with a fructose-rich diet. PTU treatment made animals hypothyroid and that resulted in total replacement of cardiac alpha-MHC with the beta-MHC isoform. Addition of fructose in the PTU diet led to reexpression of the alpha-MHC isoform to a significant level. Similar induction of alpha-MHC expression was also seen when PTU diet was combined with resveratrol, an agonist of sirtuin (SIRT) 1 deacetylase. Analysis of heart lysate of these animals indicated that fructose feeding augmented the NAD-to-NADH ratio and the cardiac SIRT1 levels, thus suggesting a role of SIRT1 in fructose-mediated activation of alpha-MHC isoform. To analyze a direct effect of SIRT1 on MHC isoform expression, we generated transgenic mice expressing SIRT1 in the heart. Treatment of these transgenic mice with PTU diet did not lead to disappearance of alpha-MHC, as it did in the nontransgenic animals. SIRT1 overexpression also activated the alpha-MHC gene promoter in transient transfection assays, thus confirming a role of SIRT1 in the induction of alpha-MHC expression. Fructose feeding also attenuated the MHC isoform shift and blocked the cardiac hypertrophy response associated with pressure overload, which was again associated with the induction of cardiac SIRT1 levels. These results demonstrate that fructose feeding protects the heart by induction of the SIRT1 deacetylase and highlight its role in the induction of alpha-MHC gene expression.
Collapse
Affiliation(s)
- Jyothish B Pillai
- Division of Cardiothoracic Surgery, Department of Surgery, Committee on Molecular Medicine and Pathology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Segal MS, Gollub E, Johnson RJ. Is the fructose index more relevant with regards to cardiovascular disease than the glycemic index? Eur J Nutr 2007; 46:406-17. [PMID: 17763967 DOI: 10.1007/s00394-007-0680-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Accepted: 08/21/2007] [Indexed: 12/17/2022]
Abstract
The glycemic index (G.I.) is a means for categorizing carbohydrates based on their ability to raise blood glucose, subsequently this index has been popularized as a way for selecting foods to reduce the risk for obesity, diabetes, and cardiovascular disease. We suggest that the G.I. is better aimed at identifying foods that stimulate insulin secretion rather than foods that stimulate insulin resistance. In this regard, fructose has a low G.I. but may be causally linked with the obesity and cardiovascular disease epidemic. The reported association of high G.I. with cardiovascular disease may be due to the association of sugar intake which contains fructose, but which has a high G.I. due to its glucose content. We propose the use of a fructose index to categorize foods and propose studies to determine the effect of low fructose diets as a means to prevent obesity, diabetes, and cardiovascular disease in the population.
Collapse
Affiliation(s)
- Mark S Segal
- Division of Nephrology, Hypertension and Transplantation, University of Florida, Gainesville, FL, USA
| | | | | |
Collapse
|
38
|
Abstract
Under normal physiological conditions, the brain utilizes only a small number of carbon sources for energy. Recently, there is growing molecular and biochemical evidence that other carbon sources, including fructose, may play a role in neuro-energetics. Fructose is the number one commercial sweetener in Western civilization with large amounts of fructose being toxic, yet fructose metabolism remains relatively poorly characterized. Fructose is purportedly metabolized via either of two pathways, the fructose-1-phosphate pathway and/or the fructose-6-phosphate pathway. Many early metabolic studies could not clearly discriminate which of these two pathways predominates, nor could they distinguish which cell types in various tissues are capable of fructose metabolism. In addition, the lack of good physiological models, the diet-induced changes in gene expression in many tissues, the involvement of multiple genes in multiple pathways involved in fructose metabolism, and the lack of characterization of some genes involved in fructose metabolism have complicated our understanding of the physiological role of fructose in neuro-energetics. A recent neuro-metabolism study of the cerebellum demonstrated fructose metabolism and co-expression of the genes specific for the fructose 1-phosphate pathway, GLUT5 (glut5) and ketohexokinase (khk), in Purkinje cells suggesting this as an active pathway in specific neurons? Meanwhile, concern over the rapid increase in dietary fructose, particularly among children, has increased awareness about how fructose is metabolized in vivo and what effects a high fructose diet might have. In this regard, establishment of cellular and molecular studies and physiological characterization of the important and/or deleterious roles fructose plays in the brain is critical. This review will discuss the status of fructose metabolism in the brain with special reference to the cerebellum and the physiological roles of the different pathways.
Collapse
Affiliation(s)
- Vincent A Funari
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
39
|
Sugimoto K, Suzuki J, Nakagawa K, Hayashi S, Enomoto T, Fujita T, Yamaji R, Inui H, Nakano Y. Eucalyptus leaf extract inhibits intestinal fructose absorption, and suppresses adiposity due to dietary sucrose in rats. Br J Nutr 2007; 93:957-63. [PMID: 16022767 DOI: 10.1079/bjn20051436] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sucrose is more lipogenic than starch, and the extreme ingestion of sucrose induces adiposity and obesity. The aim of this study was to examine the effect of the eucalyptus (Eucalyptus globulus) leaf extract (ELE) on adiposity due to dietary sucrose in rats. In addition, in this study, the effect of ELE on intestinal fructose absorption was also examined. Rats were fed a high-sucrose diet (75 % in calorie base) with or without ELE (10 g/kg diet) for 5 weeks. Body weight was lower in the rats receiving ELE than in the controls (342 (sd 37·9)v. 392 (sd 26·0) g (n7);P<0·05). Furthermore, ELE resulted in decreases in the triacylglycerol concentrations in the plasma (1·44 (sd 0·448)v.2·79 (sd 0·677) mmol/l (n7);P<0·05) and liver (19·1 (sd 5·07)v.44·1 (sd 16·28) μmol/g (n7);P<0·05). In contrast, ELE did not show any significant effects in the rats fed a starch diet. When rats were orally given ELE 10 min before fructose administration, the intestinal fructose absorption, which was examined by measuring the elevated concentration of fructose in the portal vein at 30 min after the fructose administration, was significantly inhibited in a dose-dependent manner. Furthermore, in rats fed a high-fructose diet, the plasma and hepatic triacylglycerol concentrations were significantly decreased by ELE. These results indicate that ELE, which inhibits the intestinal fructose absorption, can suppress adiposity in rats that ingest large amounts of sucrose or fructose.
Collapse
Affiliation(s)
- Keiichiro Sugimoto
- Center for Research and Develpment of Bioresources, Osaka Prefecture University, Sakai, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Asipu A, Hayward BE, O'Reilly J, Bonthron DT. Properties of normal and mutant recombinant human ketohexokinases and implications for the pathogenesis of essential fructosuria. Diabetes 2003; 52:2426-32. [PMID: 12941785 DOI: 10.2337/diabetes.52.9.2426] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alternative splicing of the ketohexokinase (fructokinase) gene generates a "central" predominantly hepatic isoform (ketohexokinase-C) and a more widely distributed ketohexokinase-A. Only the abundant hepatic isoform is known to possess activity, and no function is defined for the lower levels of ketohexokinase-A in peripheral tissues. Hepatic ketohexokinase deficiency causes the benign disorder essential fructosuria. The molecular basis of this has been defined in one family (compound heterozygosity for mutations Gly40Arg and Ala43Thr). Here we show that both ketohexokinase isoforms are indeed active. Ketohexokinase-A has much poorer substrate affinity than ketohexokinase-C for fructose but is considerably more thermostable. The Gly40Arg mutation seems null, rendering both ketohexokinase-A and ketohexokinase-C inactive and largely insoluble. The Ala43Thr mutant retains activity, but this mutation decreases the thermal stability of both ketohexokinase-A and ketohexokinase-C. At physiologic temperature, this results in significant loss of ketohexokinase-C activity but not of ketohexokinase-A. Affected individuals who carry both mutations therefore probably have a selective deficiency of hepatic ketohexokinase, with peripheral ketohexokinase-A being preserved. These findings raise the possibility that ketohexokinase-A serves an unknown physiologic function that remains intact in essential fructosuria. Further mutation analysis in this rare disorder could illuminate the question of whether ketohexokinase-A activity is, unlike that of ketohexokinase-C, physiologically indispensable.
Collapse
Affiliation(s)
- Aruna Asipu
- Molecular Medicine Unit, University of Leeds, St. James's University Hospital, Leeds, UK
| | | | | | | |
Collapse
|
41
|
Hajduch E, Litherland GJ, Turban S, Brot-Laroche E, Hundal HS. Insulin regulates the expression of the GLUT5 transporter in L6 skeletal muscle cells. FEBS Lett 2003; 549:77-82. [PMID: 12914929 DOI: 10.1016/s0014-5793(03)00773-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Skeletal muscle, a primary insulin target tissue, expresses the GLUT5 fructose transporter. Although insulin has no acute effect on GLUT5 expression and function in muscle, we show here that long-term (24 h) insulin treatment of L6 muscle cells induces a dose-dependent increase in GLUT5 protein (by up to two-fold), leading to a concomitant increase in fructose uptake. The increase in GLUT5 expression and function was suppressed by inhibitors of gene transcription and protein synthesis, suggesting that insulin promotes de novo carrier synthesis. Transfection of the GLUT5 gene promoter fused to luciferase into L6 cells revealed that insulin induced a 1.8-fold increase in GLUT5 promoter activity. Our findings indicate that insulin is capable of increasing the abundance and functional activity of GLUT5 in skeletal muscle cells and that this is most likely mediated via activation of the GLUT5 promoter.
Collapse
Affiliation(s)
- Eric Hajduch
- Division of Molecular Physiology, School of Life Sciences, MSI/WTB Complex, University of Dundee, Dundee DD1 4HN, UK
| | | | | | | | | |
Collapse
|
42
|
Abstract
PURPOSE We define the major pathways of hepatic oxalate synthesis in humans, examine the association with other metabolic pathways and identify ways that oxalate synthesis may be modified. In addition, we suggest what is required for further progress in this area. MATERIALS AND METHODS We consolidated relevant data primarily from recently published literature, considered new pharmacological approaches to decrease oxalate synthesis, and formulated an overview of the regulation and modification of oxalate synthesis pathways. RESULTS Experiments with animals, including humans, animal cells and in vitro preparations of cellular components, support the existence of a major metabolic pathway linking the amino acids serine, glycine and alanine. Oxalate synthesis is a minor, secondary reaction of a cascade of reactions termed the glyoxylate pathway, which has a prominent role in gluconeogenesis and ureagenesis. The enzymatic steps and effectors which regulate glyoxylate and oxalate synthesis are not well characterized. Pharmacological approaches can reduce oxalate synthesis by diminishing the glyoxylate pool and possibly modifying enzymatic reactions leading to glyoxylate synthesis. CONCLUSIONS The individual steps associated with glyoxylate and oxalate synthesis can be identified. The glyoxylate pathway has a significant functional role in intermediary liver metabolism but the way it is regulated is uncertain. Oxalate synthesis can be modified by drugs, indicating that primary and idiopathic hyperoxaluria may respond to pharmacological intervention.
Collapse
Affiliation(s)
- R P Holmes
- Department of Urology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | |
Collapse
|
43
|
Baker PW, Rofe AM, Bais R. Idiopathic calcium oxalate urolithiasis and endogenous oxalate production. Crit Rev Clin Lab Sci 1996; 33:39-82. [PMID: 8833627 DOI: 10.3109/10408369609101486] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Despite the great effort that has gone into investigating urolithiasis, this condition still persists as one of the major ailments of the urinary tract. Calcium oxalate urolithiasis is the most common form, accounting for some 60 to 80% of total stones. This review examines the elements (i.e., urine volume and pH and urinary excretion of calcium, oxalate, citrate, urate, magnesium, pyrophosphate, and glycosaminoglycans) that give rise to idiopathic calcium oxalate urolithiasis. Treatment strategies for idiopathic calcium oxalate urolithiasis, including lithotripsy, also are discussed. Urinary oxalate excretion is a major risk factor for calcium oxalate urolithiasis, with 85 to 95% of the urinary load derived endogenously. The factors controlling endogenous oxalate production are reviewed, including pathways for the diversion of glyoxylate from oxalate production. The use of beta-aminothiols and other substances to reduce endogenous oxalate production in subjects with idiopathic calcium oxalate urolithiasis is also discussed. A review of current methodologies for the determination of urinary oxalate is also included.
Collapse
Affiliation(s)
- P W Baker
- Institute of Medical and Veterinary Science, Adelaide, South Australia
| | | | | |
Collapse
|
44
|
Rangaswamy V, Altekar W. Ketohexokinase (ATP:D-fructose 1-phosphotransferase) from a halophilic archaebacterium, Haloarcula vallismortis: purification and properties. J Bacteriol 1994; 176:5505-12. [PMID: 8071229 PMCID: PMC196739 DOI: 10.1128/jb.176.17.5505-5512.1994] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Ketohexokinase (ATP:D-fructose 1-phosphotransferase [EC 2.7.1.3]), detected for the first time in a prokaryote, i.e., the extreme halophile Haloarcula vallismortis, was isolated and characterized from the same archaebacterium. This enzyme was characterized with respect to its molecular mass, amino acid composition, salt dependency, immunological cross-reactivity, and kinetic properties. Gel filtration and sucrose density gradient centrifugation revealed a native molecular mass of 100 kDa for halobacterial ketohexokinase, which is larger than its mammalian counterpart. The enzyme could be labeled by UV irradiation in the presence of [ gamma-32P]ATP, suggesting the involvement of a phosphoenzyme intermediate. Other catalytic features of the enzyme were similar to those of its mammalian counterparts. No antigenic cross-reactivity could be detected between the H. vallismortis ketohexokinase and the ketohexokinases from different rat tissues.
Collapse
Affiliation(s)
- V Rangaswamy
- Radiation Biology and Biochemistry Division, Bhabha Atomic Research Centre, Bombay, India
| | | |
Collapse
|
45
|
Donaldson IA, Doyle TC, Matas N. Expression of rat liver ketohexokinase in yeast results in fructose intolerance. Biochem J 1993; 291 ( Pt 1):179-86. [PMID: 8471037 PMCID: PMC1132499 DOI: 10.1042/bj2910179] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rat liver ketohexokinase (ATP:D-fructose 1-phosphotransferase; EC 2.7.1.3) was purified to homogeneity and the molecular mass of the protein was found by mass spectrometry to be 32,800 Da. The enzyme was cleaved and the amino acid sequences of seven peptides, comprising 24% of the total sequence, were determined. This sequence information was used to design oligonucleotide primers for a PCR using rat liver single-stranded cDNA as a template. The 224 bp PCR product was used as a probe to screen a rat liver cDNA library. A cDNA sequence of 1342 bp was obtained from three positive clones. This contained the entire coding region for ketohexokinase, and all seven peptides were identified in the predicted amino acid sequence. When ketohexokinase was expressed in Saccharomyces cerevisiae using the yeast expression vector pMA91, the cells became intolerant of the presence of fructose in their growth media. The growth of an exponential-phase culture was completely arrested within 90 min by the addition of fructose to a final concentration as low as 0.1% (w/v). This response is associated with an accumulation of fructose 1-phosphate. The cDNA for ketohexokinase encodes a protein composed of 299 amino acids with a combined molecular mass of 32,728 Da. This is in close agreement with the value for the isolated protein determined by mass spectrometry. The primary structure does not show any significant homology with those of other eukaryotic hexokinases, but it contains a highly conserved region that is present in three prokaryotic phosphotransferases that have furanose substrates.
Collapse
Affiliation(s)
- I A Donaldson
- Department of Biochemistry, University of Oxford, U.K
| | | | | |
Collapse
|
46
|
Abstract
A number of sulfhydryl compounds were shown to inhibit CO2 and oxalate formation from glyoxylate by rat liver homogenates and hepatocytes. The most significant inhibition occurred with cysteine and this inhibition was concentration-dependent. In rats made hyperoxaluric by administering ethylene glycol in their drinking water, daily intraperitoneal injections of cysteine caused a rapid and marked decrease in urinary oxalate excretion which was maintained over the duration of the treatment (28 days). Over this time period, the level of urinary oxalate excretion in these ethylene glycol-treated rats was reduced to that of the controls. It is postulated that the decrease is due to the formation of a cysteine-glyoxylate adduct, 2-carboxy-4-thiazolidine carboxylate, which prevents glyoxylate being further oxidized to oxalate. Cysteine or similar sulphydryl compounds may therefore have potential as therapeutic agents in the prevention of renal stones.
Collapse
Affiliation(s)
- R Bais
- Division of Clinical Chemistry, Institute of Medical and Veterinary Science, Adelaide, Australia
| | | | | |
Collapse
|
47
|
Davies DR, Detheux M, Van Schaftingen E. Fructose 1-phosphate and the regulation of glucokinase activity in isolated hepatocytes. EUROPEAN JOURNAL OF BIOCHEMISTRY 1990; 192:283-9. [PMID: 2145154 DOI: 10.1111/j.1432-1033.1990.tb19225.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fructose 1-phosphate kinase was partially purified from Clostridium difficile and used to develop specific assays of fructose 1-phosphate and fructose. The concentration of fructose 1-phosphate was below the detection limit of the assay (25 pmol/mg protein) in hepatocytes incubated in the presence of glucose as sole carbohydrate. Addition of fructose (0.05-1 mM) caused a concentration-dependent and transient increase in the fructose 1-phosphate content. Glucagon (1 microM) and ethanol (10 mM) caused a severalfold decrease in the concentration of fructose 1-phosphate in cells incubated with fructose, whereas the addition of 0.1 microM vasopressin or 10 mM glycerone, or raising the concentration of glucose from 5 mM to 20 mM had the opposite effect. All these agents caused changes in the concentration of triose phosphates that almost paralleled those of the fructose 1-phosphate concentration. Sorbitol had a similar effect to fructose in causing the formation of fructose 1-phosphate. D-Glyceraldehyde was much less potent in this respect than the ketose and its effect disappeared earlier. The effect of D-glyceraldehyde was reinforced by an increase in the glucose concentration and decreased by glucagon. Both fructose and D-glyceraldehyde stimulated the phosphorylation of glucose as estimated by the release of 3H2O from [2-3H]glucose, but the triose was less potent in this respect than fructose and its effect disappeared earlier. Glucagon and ethanol antagonised the effect of low concentrations of fructose or D-glyceraldehyde on the detritiation of glucose. These results support the proposal that fructose 1-phosphate mediates the effects of fructose, D-glyceraldehyde and sorbitol by relieving the inhibition exerted on glucokinase by a regulatory protein.
Collapse
Affiliation(s)
- D R Davies
- Laboratoire de Chimie physiologique, International Institute of Cellular and Molecular Pathology, Brussels, Belgium
| | | | | |
Collapse
|