1
|
Huang W, Jiang M, Wang X, Pan D, Chen W, Fan L. Non-Sugar Sweetener Rubusoside Alleviates Lipid Metabolism Disorder In Vivo and In Vitro by Targeting PPARγ/α, Lgals3, and Mknk2. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25601-25619. [PMID: 39508235 DOI: 10.1021/acs.jafc.4c06018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Rubusoside─a high-sweetened, nonsugar sweetener─is mainly extracted from Rubus chingii var. suavissimus (S. Lee) L. T. Lu or Rubus suavissimus S. Lee (Chinese sweet leaf tea). We previously reported that rubusoside regulates lipid metabolism disorder in Syrian golden hamsters on a high-fat diet (HFD). This study aimed to reveal the underlying mechanisms through which rubusoside alleviates lipid metabolism disorder in vivo and in vitro. First, we analyzed the therapeutic properties of rubusoside in alleviating HFD-induced lipid metabolism disorder in C57BL/6J mice. Then, we analyzed the adipogenic effect of rubusoside in normal and Lgals3/Mknk2-overexpressing 3T3-L1 cells by exploring the mechanisms on peroxisome proliferator-activated receptor-γ/α (PPARγ/α), galectin-3 (Lgals3), mitogen-activated protein kinase interacting serine/threonine kinase-2 (Mknk2), p38 mitogen-activated protein kinase (p38MAPK), and extracellular regulated protein kinases 1/2 (ERK1/2) with RT-qPCR and Western blot. Our results showed a rubusoside-mediated reduction of HFD-induced weight gain, dyslipidemia, and decelerated hepatic steatosis and adipose tissue expansion in mice as well as improved adipogenesis in 3T3-L1 cells. Mechanistically, rubusoside up-regulated the PPARγ/α expression while down-regulating Lgals3 and Mknk2 expression in vivo and in vitro. Furthermore, rubusoside attenuated the adipogenic activity of PPARγ through increasing its site-specific phosphorylation mediated by p38MAPK and ERK1/2. Taken together, our findings suggest that rubusoside alleviates lipid metabolism disorder through multiple pathways and thus holds potential for future development.
Collapse
Affiliation(s)
- Wanfang Huang
- School of Pharmacy, Guangxi University of Chinese Medicine, No. 13, Wuhe Avenue, Nanning, Guangxi 530200, China
| | - Manjing Jiang
- School of Pharmacy, Guangxi University of Chinese Medicine, No. 13, Wuhe Avenue, Nanning, Guangxi 530200, China
| | - Xue Wang
- School of Pharmacy, Guangxi University of Chinese Medicine, No. 13, Wuhe Avenue, Nanning, Guangxi 530200, China
| | - Dongjin Pan
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, No. 13, Wuhe Avenue, Nanning, Guangxi 530200, China
| | - Wenya Chen
- School of Pharmacy, Guangxi University of Chinese Medicine, No. 13, Wuhe Avenue, Nanning, Guangxi 530200, China
| | - Lanlan Fan
- School of Pharmacy, Guangxi University of Chinese Medicine, No. 13, Wuhe Avenue, Nanning, Guangxi 530200, China
| |
Collapse
|
2
|
Kawatani M, Osada H. Small-molecule inhibitors of glucose transporters. VITAMINS AND HORMONES 2024; 128:213-242. [PMID: 40097251 DOI: 10.1016/bs.vh.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Facilitative glucose transporters (GLUTs) encoded by the SLC2A genes mediate the initial steps of sugar utilization in cells. Fourteen existing GLUT family members are classified into three subclasses based on the characteristics of the gene structure. Several GLUT isoforms, especially GLUT1 and GLUT3, are overexpressed in many tumors, and their high expression correlates with poor clinical outcomes in patients. Altered energy metabolism, such as increased glycolysis, is a critical hallmark of most human cancers. Therefore, small-molecule GLUT inhibitors are promising bioprobes for understanding complex tumor metabolism and may serve as new candidate drugs for cancer therapy. Certain naturally occurring flavonoids have been shown to inhibit glucose uptake by GLUTs. Recently, a variety of potent and selective GLUT inhibitors of different chemotypes have been developed to target glycolysis-addicted tumors. Moreover, the elucidation of GLUT crystal structures has enabled high-throughput virtual screening to identify GLUT isoform-specific inhibitors. In this chapter, we provide an overview of small-molecule GLUT inhibitors, ranging from natural products to natural product-inspired and synthetic compounds.
Collapse
Affiliation(s)
- Makoto Kawatani
- Chemical Resource Development Unit, Technology Platform Division, RIKEN Center for Sustainable Resource Science (CSRS), Wako-shi, Saitama, Japan; Biomolecular Characterization Unit, Technology Platform Division, RIKEN Center for Sustainable Resource Science (CSRS), Wako-shi, Saitama, Japan.
| | - Hiroyuki Osada
- Chemical Resource Development Unit, Technology Platform Division, RIKEN Center for Sustainable Resource Science (CSRS), Wako-shi, Saitama, Japan
| |
Collapse
|
3
|
Song A, Mao Y, Wei H. GLUT5: structure, functions, diseases and potential applications. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1519-1538. [PMID: 37674366 PMCID: PMC10582729 DOI: 10.3724/abbs.2023158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/19/2023] [Indexed: 09/08/2023] Open
Abstract
Glucose transporter 5 (GLUT5) is a membrane transporter that specifically transports fructose and plays a key role in dietary fructose uptake and metabolism. In recent years, a high fructose diet has occupied an important position in the daily intake of human beings, resulting in a significant increase in the incidence of obesity and metabolic diseases worldwide. Over the past few decades, GLUT5 has been well understood to play a significant role in the pathogenesis of human digestive diseases. Recently, the role of GLUT5 in human cancer has received widespread attention, and a large number of studies have focused on exploring the effects of changes in GLUT5 expression levels on cancer cell survival, metabolism and metastasis. However, due to various difficulties and shortcomings, the molecular structure and mechanism of GLUT5 have not been fully elucidated, which to some extent prevents us from revealing the relationship between GLUT5 expression and cell carcinogenesis at the protein molecular level. In this review, we summarize the current understanding of the structure and function of mammalian GLUT5 and its relationship to intestinal diseases and cancer and suggest that GLUT5 may be an important target for cancer therapy.
Collapse
Affiliation(s)
- Aqian Song
- Department of GastroenterologyBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
| | - Yuanpeng Mao
- Department of GastroenterologyPeking University Ditan Teaching HospitalBeijing100015China
| | - Hongshan Wei
- Department of GastroenterologyBeijing Ditan HospitalCapital Medical UniversityBeijing100015China
- Department of GastroenterologyPeking University Ditan Teaching HospitalBeijing100015China
| |
Collapse
|
4
|
Suades A, Qureshi A, McComas SE, Coinçon M, Rudling A, Chatzikyriakidou Y, Landreh M, Carlsson J, Drew D. Establishing mammalian GLUT kinetics and lipid composition influences in a reconstituted-liposome system. Nat Commun 2023; 14:4070. [PMID: 37429918 DOI: 10.1038/s41467-023-39711-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/26/2023] [Indexed: 07/12/2023] Open
Abstract
Glucose transporters (GLUTs) are essential for organism-wide glucose homeostasis in mammals, and their dysfunction is associated with numerous diseases, such as diabetes and cancer. Despite structural advances, transport assays using purified GLUTs have proven to be difficult to implement, hampering deeper mechanistic insights. Here, we have optimized a transport assay in liposomes for the fructose-specific isoform GLUT5. By combining lipidomic analysis with native MS and thermal-shift assays, we replicate the GLUT5 transport activities seen in crude lipids using a small number of synthetic lipids. We conclude that GLUT5 is only active under a specific range of membrane fluidity, and that human GLUT1-4 prefers a similar lipid composition to GLUT5. Although GLUT3 is designated as the high-affinity glucose transporter, in vitro D-glucose kinetics demonstrates that GLUT1 and GLUT3 actually have a similar KM, but GLUT3 has a higher turnover. Interestingly, GLUT4 has a high KM for D-glucose and yet a very slow turnover, which may have evolved to ensure uptake regulation by insulin-dependent trafficking. Overall, we outline a much-needed transport assay for measuring GLUT kinetics and our analysis implies that high-levels of free fatty acid in membranes, as found in those suffering from metabolic disorders, could directly impair glucose uptake.
Collapse
Affiliation(s)
- Albert Suades
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16c, SE-106 91, Stockholm, Sweden
| | - Aziz Qureshi
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16c, SE-106 91, Stockholm, Sweden
| | - Sarah E McComas
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16c, SE-106 91, Stockholm, Sweden
| | - Mathieu Coinçon
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16c, SE-106 91, Stockholm, Sweden
| | - Axel Rudling
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24, Uppsala, Sweden
| | - Yurie Chatzikyriakidou
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16c, SE-106 91, Stockholm, Sweden
| | - Michael Landreh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, SE-171 65, Solna, Sweden
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24, Uppsala, Sweden
| | - David Drew
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius v. 16c, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
5
|
Chamarthy S, Mekala JR. Functional importance of glucose transporters and chromatin epigenetic factors in Glioblastoma Multiforme (GBM): possible therapeutics. Metab Brain Dis 2023; 38:1441-1469. [PMID: 37093461 DOI: 10.1007/s11011-023-01207-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/22/2023] [Indexed: 04/25/2023]
Abstract
Glioblastoma Multiforme (GBM) is an aggressive brain cancer affecting glial cells and is chemo- and radio-resistant. Glucose is considered the most vital energy source for cancer cell proliferation. During metabolism, hexose molecules will be transported into the cells via transmembrane proteins known as glucose transporter (GLUT). Among them, GLUT-1 and GLUT-3 play pivotal roles in glucose transport in GBM. Knockdown studies have established the role of GLUT-1, and GLUT-3 mediated glucose transport in GBM cells, providing insight into GLUT-mediated cancer signaling and cancer aggressiveness. This review focussed on the vital role of GLUT-1 and GLUT-3 proteins, which regulate glucose transport. Recent studies have identified the role of GLUT inhibitors in effective cancer prevention. Several of them are in clinical trials. Understanding and functional approaches towards glucose-mediated cell metabolism and chromatin epigenetics will provide valuable insights into the mechanism of cancer aggressiveness, cancer stemness, and chemo-resistance in Glioblastoma Multiforme (GBM). This review summarizes the role of GLUT inhibitors, micro-RNAs, and long non-coding RNAs that aid in inhibiting glucose uptake by the GBM cells and other cancer cells leading to the identification of potential therapeutic, prognostic as well as diagnostic markers. Furthermore, the involvement of epigenetic factors, such as microRNAs, in regulating glycolytic genes was demonstrated.
Collapse
Affiliation(s)
- Sahiti Chamarthy
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Green Fields, Vaddeswaram, Guntur, Andhra Pradesh, 522302, India
| | - Janaki Ramaiah Mekala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Green Fields, Vaddeswaram, Guntur, Andhra Pradesh, 522302, India.
| |
Collapse
|
6
|
Temre MK, Kumar A, Singh SM. An appraisal of the current status of inhibition of glucose transporters as an emerging antineoplastic approach: Promising potential of new pan-GLUT inhibitors. Front Pharmacol 2022; 13:1035510. [PMID: 36386187 PMCID: PMC9663470 DOI: 10.3389/fphar.2022.1035510] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/18/2022] [Indexed: 07/23/2023] Open
Abstract
Neoplastic cells displayed altered metabolism with accelerated glycolysis. Therefore, these cells need a mammoth supply of glucose for which they display an upregulated expression of various glucose transporters (GLUT). Thus, novel antineoplastic strategies focus on inhibiting GLUT to intersect the glycolytic lifeline of cancer cells. This review focuses on the current status of various GLUT inhibition scenarios. The GLUT inhibitors belong to both natural and synthetic small inhibitory molecules category. As neoplastic cells express multiple GLUT isoforms, it is necessary to use pan-GLUT inhibitors. Nevertheless, it is also necessary that such pan-GLUT inhibitors exert their action at a low concentration so that normal healthy cells are left unharmed and minimal injury is caused to the other vital organs and systems of the body. Moreover, approaches are also emerging from combining GLUT inhibitors with other chemotherapeutic agents to potentiate the antineoplastic action. A new pan-GLUT inhibitor named glutor, a piperazine-one derivative, has shown a potent antineoplastic action owing to its inhibitory action exerted at nanomolar concentrations. The review discusses the merits and limitations of the existing GLUT inhibitory approach with possible future outcomes.
Collapse
Affiliation(s)
- Mithlesh Kumar Temre
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ajay Kumar
- Deparment of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sukh Mahendra Singh
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
7
|
Targeting Glucose Metabolism Enzymes in Cancer Treatment: Current and Emerging Strategies. Cancers (Basel) 2022; 14:cancers14194568. [PMID: 36230492 PMCID: PMC9559313 DOI: 10.3390/cancers14194568] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Reprogramming of glucose metabolism is a hallmark of cancer and can be targeted by therapeutic agents. Some metabolism regulators, such as ivosidenib and enasidenib, have been approved for cancer treatment. Currently, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Furthermore, some natural products have shown efficacy in killing tumor cells by regulating glucose metabolism, offering novel therapeutic opportunities in cancer. However, most of them have failed to be translated into clinical applications due to low selectivity, high toxicity, and side effects. Recent studies suggest that combining glucose metabolism modulators with chemotherapeutic drugs, immunotherapeutic drugs, and other conventional anticancer drugs may be a future direction for cancer treatment. Abstract Reprogramming of glucose metabolism provides sufficient energy and raw materials for the proliferation, metastasis, and immune escape of cancer cells, which is enabled by glucose metabolism-related enzymes that are abundantly expressed in a broad range of cancers. Therefore, targeting glucose metabolism enzymes has emerged as a promising strategy for anticancer drug development. Although several glucose metabolism modulators have been approved for cancer treatment in recent years, some limitations exist, such as a short half-life, poor solubility, and numerous adverse effects. With the rapid development of medicinal chemicals, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Additionally, several studies have found that some natural products can suppress cancer progression by regulating glucose metabolism enzymes. In this review, we summarize the mechanisms underlying the reprogramming of glucose metabolism and present enzymes that could serve as therapeutic targets. In addition, we systematically review the existing drugs targeting glucose metabolism enzymes, including small-molecule modulators and natural products. Finally, the opportunities and challenges for glucose metabolism enzyme-targeted anticancer drugs are also discussed. In conclusion, combining glucose metabolism modulators with conventional anticancer drugs may be a promising cancer treatment strategy.
Collapse
|
8
|
Fang XY, Qi LW, Chen HF, Gao P, Zhang Q, Leng RX, Fan YG, Li BZ, Pan HF, Ye DQ. The Interaction Between Dietary Fructose and Gut Microbiota in Hyperuricemia and Gout. Front Nutr 2022; 9:890730. [PMID: 35811965 PMCID: PMC9257186 DOI: 10.3389/fnut.2022.890730] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
With the worldwide epidemics of hyperuricemia and associated gout, the diseases with purine metabolic disorders have become a serious threat to human public health. Accumulating evidence has shown that they have been linked to increased consumption of fructose in humans, we hereby made a timely review on the roles of fructose intake and the gut microbiota in regulating purine metabolism, together with the potential mechanisms by which excessive fructose intake contributes to hyperuricemia and gout. To this end, we focus on the understanding of the interaction between a fructose-rich diet and the gut microbiota in hyperuricemia and gout to seek for safe, cheap, and side-effect-free clinical interventions. Furthermore, fructose intake recommendations for hyperuricemia and gout patients, as well as the variety of probiotics and prebiotics with uric acid-lowering effects targeting the intestinal tract are also summarized to provide reference and guidance for the further research.
Collapse
Affiliation(s)
- Xin-yu Fang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Liang-wei Qi
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Hai-feng Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Peng Gao
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Qin Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Rui-xue Leng
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Yin-guang Fan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Bao-zhu Li
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Hai-feng Pan
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
| | - Dong-qing Ye
- Department of Epidemiology and Health Statistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui, Hefei, China
- *Correspondence: Dong-qing Ye
| |
Collapse
|
9
|
Rana N, Aziz MA, Oraby AK, Wuest M, Dufour J, Abouzid KAM, Wuest F, West FG. Towards Selective Binding to the GLUT5 Transporter: Synthesis, Molecular Dynamics and In Vitro Evaluation of Novel C-3-Modified 2,5-Anhydro-D-mannitol Analogs. Pharmaceutics 2022; 14:828. [PMID: 35456662 PMCID: PMC9032776 DOI: 10.3390/pharmaceutics14040828] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/27/2022] [Accepted: 04/07/2022] [Indexed: 02/03/2023] Open
Abstract
Deregulation and changes in energy metabolism are emergent and important biomarkers of cancer cells. The uptake of hexoses in cancer cells is mediated by a family of facilitative hexose membrane-transporter proteins known as Glucose Transporters (GLUTs). In the clinic, numerous breast cancers do not show elevated glucose metabolism (which is mediated mainly through the GLUT1 transporter) and may use fructose as an alternative energy source. The principal fructose transporter in most cancer cells is GLUT5, and its mRNA was shown to be elevated in human breast cancer. This offers an alternative strategy for early detection using fructose analogs. In order to selectively scout GLUT5 binding-pocket requirements, we designed, synthesized and screened a new class of fructose mimics based upon the 2,5-anhydromannitol scaffold. Several of these compounds display low millimolar IC50 values against the known high-affinity 18F-labeled fructose-based probe 6-deoxy-6-fluoro-D-fructose (6-FDF) in murine EMT6 breast cancer cells. In addition, this work used molecular docking and molecular dynamics simulations (MD) with previously reported GLUT5 structures to gain better insight into hexose-GLUT interactions with selected ligands governing their preference for GLUT5 compared to other GLUTs. The improved inhibition of these compounds, and the refined model for their binding, set the stage for the development of high-affinity molecular imaging probes targeting cancers that express the GLUT5 biomarker.
Collapse
Affiliation(s)
- Natasha Rana
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
- Department of Oncology, University of Alberta-Cross Cancer Institute, Edmonton, AB T6G IZ2, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, 2-132 Li Ka Shing, Edmonton, AB T6G 2E1, Canada
| | - Marwa A Aziz
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo P.O. Box 11566, Egypt
| | - Ahmed K Oraby
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Misr University of Science & Technology, Al-Motamayez District, 6th of October City P.O. Box 77, Egypt
| | - Melinda Wuest
- Department of Oncology, University of Alberta-Cross Cancer Institute, Edmonton, AB T6G IZ2, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, 2-132 Li Ka Shing, Edmonton, AB T6G 2E1, Canada
| | - Jennifer Dufour
- Department of Oncology, University of Alberta-Cross Cancer Institute, Edmonton, AB T6G IZ2, Canada
| | - Khaled A M Abouzid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo P.O. Box 11566, Egypt
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Sadat City P.O. Box 32897, Egypt
| | - Frank Wuest
- Department of Oncology, University of Alberta-Cross Cancer Institute, Edmonton, AB T6G IZ2, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, 2-132 Li Ka Shing, Edmonton, AB T6G 2E1, Canada
| | - F G West
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, 2-132 Li Ka Shing, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
10
|
Xia J, Ma S, Zhu X, Chen C, Zhang R, Cao Z, Chen X, Zhang L, Zhu Y, Zhang S, Li S, Gu G, Wei X, Yu K, Wang J. Versatile ginsenoside Rg3 liposomes inhibit tumor metastasis by capturing circulating tumor cells and destroying metastatic niches. SCIENCE ADVANCES 2022; 8:eabj1262. [PMID: 35148178 PMCID: PMC8836824 DOI: 10.1126/sciadv.abj1262] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Limited circulating tumor cells (CTCs) capturing efficiency and lack of regulation capability on CTC-supportive metastatic niches (MNs) are two main obstacles hampering the clinical translation of conventional liposomes for the treatment of metastatic breast cancers. Traditional delivery strategies, such as ligand modification and immune modulator co-encapsulation for nanocarriers, are inefficient and laborious. Here, a multifunctional Rg3 liposome loading with docetaxel (Rg3-Lp/DTX) was developed, in which Rg3 was proved to intersperse in the phospholipid bilayer and exposed its glycosyl on the liposome surface. Therefore, it exhibited much higher CTC-capturing efficiency via interaction with glucose transporter 1 (Glut1) overexpressed on CTCs. After reaching the lungs with CTCs, Rg3 inhibited the formation of MNs by reversing the immunosuppressive microenvironment. Together, Rg3-Lp/DTX exhibited excellent metastasis inhibition capacity by CTC ("seeds") neutralization and MN ("soil") inhibition. The strategy has great clinical translation prospects for antimetastasis treatment with enhanced therapeutic efficacy and simple preparation process.
Collapse
Affiliation(s)
- Jiaxuan Xia
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Shaojie Ma
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430071, China
| | - Xi Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chen Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Ru Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Zhonglian Cao
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Xing Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Longlong Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Ying Zhu
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Shuya Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Shiyi Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Guolong Gu
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Xunbin Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Kunqian Yu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Institutes of Integrative Medicine, Fudan University, Shanghai 201203, China
- Corresponding author.
| |
Collapse
|
11
|
Rubusoside relieves lipopolysaccharide-induced acute lung injury via modulating inflammatory responses: in vitro and in vivo studies. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-021-02246-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
GLUT3 inhibitor discovery through in silico ligand screening and in vivo validation in eukaryotic expression systems. Sci Rep 2022; 12:1429. [PMID: 35082341 PMCID: PMC8791944 DOI: 10.1038/s41598-022-05383-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/06/2022] [Indexed: 12/30/2022] Open
Abstract
The passive transport of glucose and related hexoses in human cells is facilitated by members of the glucose transporter family (GLUT, SLC2 gene family). GLUT3 is a high-affinity glucose transporter primarily responsible for glucose entry in neurons. Changes in its expression have been implicated in neurodegenerative diseases and cancer. GLUT3 inhibitors can provide new ways to probe the pathophysiological role of GLUT3 and tackle GLUT3-dependent cancers. Through in silico screening of an ~ 8 million compounds library against the inward- and outward-facing models of GLUT3, we selected ~ 200 ligand candidates. These were tested for in vivo inhibition of GLUT3 expressed in hexose transporter-deficient yeast cells, resulting in six new GLUT3 inhibitors. Examining their specificity for GLUT1-5 revealed that the most potent GLUT3 inhibitor (G3iA, IC50 ~ 7 µM) was most selective for GLUT3, inhibiting less strongly only GLUT2 (IC50 ~ 29 µM). None of the GLUT3 inhibitors affected GLUT5, three inhibited GLUT1 with equal or twofold lower potency, and four showed comparable or two- to fivefold better inhibition of GLUT4. G3iD was a pan-Class 1 GLUT inhibitor with the highest preference for GLUT4 (IC50 ~ 3.9 µM). Given the prevalence of GLUT1 and GLUT3 overexpression in many cancers and multiple myeloma’s reliance on GLUT4, these GLUT3 inhibitors may discriminately hinder glucose entry into various cancer cells, promising novel therapeutic avenues in oncology.
Collapse
|
13
|
Oenothein B in Eucalyptus Leaf Extract Suppresses Fructose Absorption in Caco-2 Cells. Molecules 2021; 27:molecules27010122. [PMID: 35011353 PMCID: PMC8746427 DOI: 10.3390/molecules27010122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/10/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022] Open
Abstract
Inhibition of fructose absorption may suppress adiposity and adiposity-related diseases caused by fructose ingestion. Eucalyptus leaf extract (ELE) inhibits intestinal fructose absorption (but not glucose absorption); however, its active compound has not yet been identified. Therefore, we evaluated the inhibitory activity of ELE obtained from Eucalyptus globulus using an intestinal fructose permeation assay with the human intestinal epithelial cell line Caco-2. The luminal sides of a cell monolayer model cultured on membrane filters were exposed to fructose with or without the ELE. Cellular fructose permeation was evaluated by measuring the fructose concentration in the medium on the basolateral side. ELE inhibited 65% of fructose absorption at a final concentration of 1 mg/mL. Oenothein B isolated from the ELE strongly inhibited fructose absorption; the inhibition rate was 63% at a final concentration of 5 μg/mL. Oenothein B did not affect glucose absorption. In contrast, the other major constituents (i.e., gallic acid and ellagic acid) showed little fructose-inhibitory activity. To our knowledge, this is the first report that oenothein B in ELE strongly inhibits fructose absorption in vitro. ELE containing oenothein B can prevent and ameliorate obesity and other diseases caused by dietary fructose consumption.
Collapse
|
14
|
Ko JA, Kim SY, Ahn HS, Go JG, Ryu YB, Lee WS, Wee YJ, Park JS, Kim D, Kim YM. Characterization of a lactic acid bacterium-derived β-glucosidase for the production of rubusoside from stevioside. Enzyme Microb Technol 2021; 153:109939. [PMID: 34798448 DOI: 10.1016/j.enzmictec.2021.109939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/26/2022]
Abstract
Rubusoside, which is used as a natural sweetener or a solubilizing agent for water-insoluble functional materials, is currently expensive to produce owing to the high cost of the membrane-based technologies needed for its extraction and purification from the sweet tea plant (Rubus suavissimus S. Lee). Therefore, this study was carried out to screen for lactic acid bacteria that possess enzymes capable of bio-transforming stevioside into rubusoside. Subsequently, one such rubusoside-producing enzyme was isolated from Lactobacillus plantarum GS100. Located on the bacterial cell surface, this enzyme was stable at pH 4.5-6.5 and 30-40 °C, and it produced rubusoside as a major product through its stevioside-hydrolyzing activity. Importantly, the enzyme showed higher β-glucosidase activity toward the β-linked glucosidic bond of stevioside than toward other β-linked glucobioses. Under optimal conditions, 70 U/L of the rubusoside-producing enzyme could produce 69.03 mM rubusoside from 190 mM stevioside. The β-glucosidase activity on the cell surface was high at 35 h of culture. This is the first report detailing the production of rubusoside from stevioside by an enzyme derived from a food-grade lactic acid bacterium. The application of this β-glucosidase could greatly reduce the cost of rubusoside production, hence benefiting all industries that use this natural product.
Collapse
Affiliation(s)
- Jin-A Ko
- Department of Food Science & Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - So-Yeon Kim
- Department of Food Science & Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hye-Soo Ahn
- Department of Food Science & Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jae-Gyune Go
- Department of Food Science & Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Young-Bae Ryu
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeollabuk-do 56212, Republic of Korea
| | - Woo Song Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeollabuk-do 56212, Republic of Korea
| | - Young-Jung Wee
- Department of Food Science and Technology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jun-Seong Park
- Department of Engineering Chemistry, Chungbuk National University, Chongju 28644, Republic of Korea
| | - Doman Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, Republic of Korea
| | - Young-Min Kim
- Department of Food Science & Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
15
|
Mao Y, Chen Z, Ren Y, Sun Y, Wang Y. Whole-Cell Biocatalyst for Rubusoside Production in Saccharomyces cerevisiae. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:13155-13163. [PMID: 34699718 DOI: 10.1021/acs.jafc.1c04873] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Rubusoside (Rub) is a highly sweet diterpene glycoside mainly isolated from the leaves of Rubus suavissimus (Rosaceae). It has been used as a low-calorie natural sweetener for decades and was recently found to be a potential drug lead. In this study, we designed a whole-cell biocatalyst to achieve the glycosylation of steviol to Rub in Saccharomyces cerevisiae. The sucrose synthases were applied to construct a uridine diphosphate glucose regeneration system, which were coupled with optimal combinations of different uridine diphosphate (UDP) glycosyltransferases from multiple plant species. After optimization of reaction conditions, the residues in SrUGT74G1 probably influencing glycosylation efficiency were subjected to site-directed mutagenesis. Double mutations of S84A/E87A reduced the accumulation of intermediates, finally glucosylating 1.27 g/L steviol to 0.45 ± 0.06 g/L steviolmonoside (conversion rate = 23.3%) and 1.92 ± 0.17 g/L Rub (conversion rate = 74.9%). A high efficiency of Rub biosynthesis could be achieved without supply of additional UDPG. This work provided the first example of multi-step glycosylation reactions in whole-cell biocatalysis, which laid a foundation of scalable production of the value-added diterpene sweetener in the future.
Collapse
Affiliation(s)
- Yaping Mao
- East China University of Science and Technology, Shanghai 200237, China
- University of Chinese Academy of Sciences, Beijing 100039, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Zhuo Chen
- University of Chinese Academy of Sciences, Beijing 100039, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yuhong Ren
- East China University of Science and Technology, Shanghai 200237, China
| | - Yuwei Sun
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yong Wang
- East China University of Science and Technology, Shanghai 200237, China
- CAS-Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
16
|
Identification of new GLUT2-selective inhibitors through in silico ligand screening and validation in eukaryotic expression systems. Sci Rep 2021; 11:13751. [PMID: 34215797 PMCID: PMC8253845 DOI: 10.1038/s41598-021-93063-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/14/2021] [Indexed: 01/07/2023] Open
Abstract
Glucose is an essential energy source for cells. In humans, its passive diffusion through the cell membrane is facilitated by members of the glucose transporter family (GLUT, SLC2 gene family). GLUT2 transports both glucose and fructose with low affinity and plays a critical role in glucose sensing mechanisms. Alterations in the function or expression of GLUT2 are involved in the Fanconi-Bickel syndrome, diabetes, and cancer. Distinguishing GLUT2 transport in tissues where other GLUTs coexist is challenging due to the low affinity of GLUT2 for glucose and fructose and the scarcity of GLUT-specific modulators. By combining in silico ligand screening of an inward-facing conformation model of GLUT2 and glucose uptake assays in a hexose transporter-deficient yeast strain, in which the GLUT1-5 can be expressed individually, we identified eleven new GLUT2 inhibitors (IC50 ranging from 0.61 to 19.3 µM). Among them, nine were GLUT2-selective, one inhibited GLUT1-4 (pan-Class I GLUT inhibitor), and another inhibited GLUT5 only. All these inhibitors dock to the substrate cavity periphery, close to the large cytosolic loop connecting the two transporter halves, outside the substrate-binding site. The GLUT2 inhibitors described here have various applications; GLUT2-specific inhibitors can serve as tools to examine the pathophysiological role of GLUT2 relative to other GLUTs, the pan-Class I GLUT inhibitor can block glucose entry in cancer cells, and the GLUT2/GLUT5 inhibitor can reduce the intestinal absorption of fructose to combat the harmful effects of a high-fructose diet.
Collapse
|
17
|
Hu Z, Liu X, Tian M, Ma Y, Jin B, Gao W, Cui G, Guo J, Huang L. Recent progress and new perspectives for diterpenoid biosynthesis in medicinal plants. Med Res Rev 2021; 41:2971-2997. [PMID: 33938025 DOI: 10.1002/med.21816] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 12/25/2022]
Abstract
Diterpenoids, including more than 18,000 compounds, represent an important class of metabolites that encompass both phytohormones and some industrially relevant compounds. These molecules with complex, diverse structures and physiological activities, have high value in the pharmaceutical industry. Most medicinal diterpenoids are extracted from plants. Major advances in understanding the biosynthetic pathways of these active compounds are providing unprecedented opportunities for the industrial production of diterpenoids by metabolic engineering and synthetic biology. Here, we summarize recent developments in the field of diterpenoid biosynthesis from medicinal herbs. An overview of the pathways and known biosynthetic enzymes is presented. In particular, we look at the main findings from the past decade and review recent progress in the biosynthesis of different groups of ringed compounds. We also discuss diterpenoid production using synthetic biology and metabolic engineering strategies, and draw on new technologies and discoveries to bring together many components into a useful framework for diterpenoid production.
Collapse
Affiliation(s)
- Zhimin Hu
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiuyu Liu
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,School of Pharmaceutical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Mei Tian
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Ma
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baolong Jin
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Gao
- School of Pharmaceutical, Sciences, Capital Medical University, Beijing, China
| | - Guanghong Cui
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Juan Guo
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Li F, Egea PF, Vecchio AJ, Asial I, Gupta M, Paulino J, Bajaj R, Dickinson MS, Ferguson-Miller S, Monk BC, Stroud RM. Highlighting membrane protein structure and function: A celebration of the Protein Data Bank. J Biol Chem 2021; 296:100557. [PMID: 33744283 PMCID: PMC8102919 DOI: 10.1016/j.jbc.2021.100557] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/10/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Biological membranes define the boundaries of cells and compartmentalize the chemical and physical processes required for life. Many biological processes are carried out by proteins embedded in or associated with such membranes. Determination of membrane protein (MP) structures at atomic or near-atomic resolution plays a vital role in elucidating their structural and functional impact in biology. This endeavor has determined 1198 unique MP structures as of early 2021. The value of these structures is expanded greatly by deposition of their three-dimensional (3D) coordinates into the Protein Data Bank (PDB) after the first atomic MP structure was elucidated in 1985. Since then, free access to MP structures facilitates broader and deeper understanding of MPs, which provides crucial new insights into their biological functions. Here we highlight the structural and functional biology of representative MPs and landmarks in the evolution of new technologies, with insights into key developments influenced by the PDB in magnifying their impact.
Collapse
Affiliation(s)
- Fei Li
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA; Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Pascal F Egea
- Department of Biological Chemistry, School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Alex J Vecchio
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | | | - Meghna Gupta
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Joana Paulino
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Ruchika Bajaj
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Miles Sasha Dickinson
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Shelagh Ferguson-Miller
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Brian C Monk
- Sir John Walsh Research Institute and Department of Oral Sciences, University of Otago, North Dunedin, Dunedin, New Zealand
| | - Robert M Stroud
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
19
|
Tilekar K, Upadhyay N, Iancu CV, Pokrovsky V, Choe JY, Ramaa CS. Power of two: combination of therapeutic approaches involving glucose transporter (GLUT) inhibitors to combat cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188457. [PMID: 33096154 PMCID: PMC7704680 DOI: 10.1016/j.bbcan.2020.188457] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
Cancer research of the Warburg effect, a hallmark metabolic alteration in tumors, focused attention on glucose metabolism whose targeting uncovered several agents with promising anticancer effects at the preclinical level. These agents' monotherapy points to their potential as adjuvant combination therapy to existing standard chemotherapy in human trials. Accordingly, several studies on combining glucose transporter (GLUT) inhibitors with chemotherapeutic agents, such as doxorubicin, paclitaxel, and cytarabine, showed synergistic or additive anticancer effects, reduced chemo-, radio-, and immuno-resistance, and reduced toxicity due to lowering the therapeutic doses required for desired chemotherapeutic effects, as compared with monotherapy. The combinations have been specifically effective in treating cancer glycolytic phenotypes, such as pancreatic and breast cancers. Even combining GLUT inhibitors with other glycolytic inhibitors and energy restriction mimetics seems worthwhile. Though combination clinical trials are in the early phase, initial results are intriguing. The various types of GLUTs, their role in cancer progression, GLUT inhibitors, and their anticancer mechanism of action have been reviewed several times. However, utilizing GLUT inhibitors as combination therapeutics has received little attention. We consider GLUT inhibitors agents that directly affect glucose transporters by binding to them or indirectly alter glucose transport by changing the transporters' expression level. This review mainly focuses on summarizing the effects of various combinations of GLUT inhibitors with other anticancer agents and providing a perspective on the current status.
Collapse
Affiliation(s)
- Kalpana Tilekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Neha Upadhyay
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Cristina V. Iancu
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - Vadim Pokrovsky
- Laboratory of Combined Therapy, N.N. Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People’s Friendship University, Moscow, Russia
| | - Jun-yong Choe
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - C. S. Ramaa
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| |
Collapse
|
20
|
Holman GD. Structure, function and regulation of mammalian glucose transporters of the SLC2 family. Pflugers Arch 2020; 472:1155-1175. [PMID: 32591905 PMCID: PMC7462842 DOI: 10.1007/s00424-020-02411-3] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
The SLC2 genes code for a family of GLUT proteins that are part of the major facilitator superfamily (MFS) of membrane transporters. Crystal structures have recently revealed how the unique protein fold of these proteins enables the catalysis of transport. The proteins have 12 transmembrane spans built from a replicated trimer substructure. This enables 4 trimer substructures to move relative to each other, and thereby alternately opening and closing a cleft to either the internal or the external side of the membrane. The physiological substrate for the GLUTs is usually a hexose but substrates for GLUTs can include urate, dehydro-ascorbate and myo-inositol. The GLUT proteins have varied physiological functions that are related to their principal substrates, the cell type in which the GLUTs are expressed and the extent to which the proteins are associated with subcellular compartments. Some of the GLUT proteins translocate between subcellular compartments and this facilitates the control of their function over long- and short-time scales. The control of GLUT function is necessary for a regulated supply of metabolites (mainly glucose) to tissues. Pathophysiological abnormalities in GLUT proteins are responsible for, or associated with, clinical problems including type 2 diabetes and cancer and a range of tissue disorders, related to tissue-specific GLUT protein profiles. The availability of GLUT crystal structures has facilitated the search for inhibitors and substrates and that are specific for each GLUT and that can be used therapeutically. Recent studies are starting to unravel the drug targetable properties of each of the GLUT proteins.
Collapse
Affiliation(s)
- Geoffrey D Holman
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK.
| |
Collapse
|
21
|
Development of a Multifunction Set Yogurt Using Rubus suavissimus S. Lee (Chinese Sweet Tea) Extract. Foods 2020; 9:foods9091163. [PMID: 32846883 PMCID: PMC7555928 DOI: 10.3390/foods9091163] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/16/2020] [Accepted: 08/21/2020] [Indexed: 01/24/2023] Open
Abstract
Rubus suavissimus S. Lee leaves, also known as Chinese sweet tea or Tiancha, are used in folk medicine in southern China. This study evaluated the impact of the addition of Chinese sweet tea extract (0.25%, 0.5%, and 1%) on the chemical composition, organoleptic properties, yogurt culture viability, and biological activities (i.e., antioxidant, anticancer, and antihypertensive activities) of yogurt. Seven phenolic compounds were reported in Chinese sweet tea for the first time. The numbers of the yogurt culture were similar across all yogurt treatments. The yogurt supernatant with 0.25%, 0.5%, and 1% Chinese sweet tea extract had a total phenolic content that was 3.6-, 6.1-, and 11.2-fold higher, respectively, than that of the control yogurt. The biological activities were significantly increased by the addition of Chinese sweet tea extract: Yogurt with the addition of 1% Chinese sweet tea extract had the highest biological activities in terms of the antioxidant activity (92.43%), antihypertensive activity (82.03%), and inhibition of the Caco-2 cell line (67.46%). Yogurt with the addition of 0.5% Chinese sweet tea extract received the highest aroma and overall acceptability scores. Overall, Chinese sweet tea extract is a promising food ingredient for producing functional yogurt products that may substantially contribute to reducing the risk of developing chronic diseases such as cancer and cardiovascular disease.
Collapse
|
22
|
Permuted 2,4-thiazolidinedione (TZD) analogs as GLUT inhibitors and their in-vitro evaluation in leukemic cells. Eur J Pharm Sci 2020; 154:105512. [PMID: 32801003 DOI: 10.1016/j.ejps.2020.105512] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/24/2020] [Accepted: 08/10/2020] [Indexed: 01/04/2023]
Abstract
Cancer is a heterogeneous disease, and its treatment requires the identification of new ways to thwart tumor cells. Amongst such emerging targets are glucose transporters (GLUTs, SLC2 family), which are overexpressed by almost all types of cancer cells; their inhibition provides a strategy to disrupt tumor metabolism selectively, leading to antitumor effects. Here, novel thiazolidinedione (TZD) derivatives were designed, synthesized, characterized, and evaluated for their GLUT1, GLUT4, and GLUT5 inhibitory potential, followed by in-vitro cytotoxicity determination in leukemic cell lines. Compounds G5, G16, and G17 inhibited GLUT1, with IC50 values of 5.4 ± 1.3, 26.6 ± 1.8, and 12.6 ± 1.2 μM, respectively. G17 was specific for GLUT1, G16 inhibited GLUT4 (IC50 = 21.6 ± 4.5 μM) comparably but did not affect GLUT5. The most active compound, G5, inhibited all three GLUT types, with GLUT4 IC50 = 9.5 ± 2.8 μM, and GLUT5 IC50 = 34.5 ± 2.4 μM. Docking G5, G16, and G17 to the inward- and outward-facing structural models of GLUT1 predicted ligand binding affinities consistent with the kinetic inhibition data and implicated E380 and W388 of GLUT1 vs. their substitutions in GLUT5 (A388 and A396, respectively) in inhibitor preference for GLUT1. G5 inhibited the proliferation of leukemia CEM cells at low micromolar range (IC50 = 13.4 μM) while being safer for normal blood cells. Investigation of CEM cell cycle progression after treatment with G5 showed that cells accumulated in the G2/M phase. Flow cytometric apoptosis studies revealed that compound G5 induced both early and late-stage apoptosis in CEM cells.
Collapse
|
23
|
Zhao L, Wang Y, Li Z, Wang X, Chen Y, Wu X. Enzymatic Monoglucosylation of Rubusoside and the Structure-Sweetness/Taste Relationship of Monoglucosyl Derivatives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8702-8709. [PMID: 32686405 DOI: 10.1021/acs.jafc.0c03236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Monoglucosylation of rubusoside not only could increase its structural diversity but may also improve its taste. To biosynthesize the monoglucosyl rubusosides, a series of glycosyltransferases and glycosynthases were screened to identify the enzymes capable of specifically glycosylating the hydroxyl groups of the 13-O-β-d-glucosyl and 19-COO-β-d-glucosyl moieties. After structural characterization, the effect of structure on sweetness and taste was established based on these rubusoside-derived analogues, including two first characterized compounds. β-Monoglucosylation of two 2-hydroxyl groups, as well as α-monoglucosylations of the 4- and 6-hydroxyl groups of the 13-glucosyl moiety, could significantly increase the relative sweetness of rubusoside to 140 while maintaining or improving the taste quality. In contrast, monoglucosylations of other hydroxyl groups in our study usually decreased the taste quality of the rubusoside. Additionally, the possibility of a negative influence of these monoglucosylated derivatives on the function of islets was preliminarily excluded, which should facilitate the development of rubusoside-derived sweeteners.
Collapse
Affiliation(s)
- Ling Zhao
- Laboratory of Chemical Biology, College of Life Sciences and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu Province 211198, PR China
| | - Yao Wang
- Laboratory of Chemical Biology, College of Life Sciences and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu Province 211198, PR China
| | - Zhenlin Li
- Department of Pharmaceutical Analysis and Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, 100 Shizi St. Hongshan Rd. Nanjing, Jiangsu Province 210028, PR China
| | - Xiaonan Wang
- Department of Biochemistry, College of Life Sciences and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu Province 211198, PR China
| | - Yijun Chen
- Laboratory of Chemical Biology, College of Life Sciences and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu Province 211198, PR China
| | - Xuri Wu
- Department of Biochemistry, College of Life Sciences and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu Province 211198, PR China
| |
Collapse
|
24
|
Wang C, Tang J, Qian B, Zeng Z, Gao Y, Song JL. Rubusoside alleviates the ovalbumin-induced mice allergic asthma by modulating the NF-κB activation. J Food Biochem 2020; 44:e13187. [PMID: 32185800 DOI: 10.1111/jfbc.13187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/20/2022]
Abstract
The anti-inflammatory and anti-asthmatic effects of rubusoside (Rbs) were investigated in the ovalbumin (OVA)-induced asthmatic mice, followed by effective attenuation of Rbs treatment on the airway hyperresponsiveness and reduction of inflammatory cells inside the bronchoalveolar lavage fluid (BALF). The mitigation of inflammatory infiltration as a result of Rbs treatment was histologically observed in these mice lungs. Rbs contributed to the decrease of inflammatory cytokines (TNF-α, IL-13, IL-6, IL-5, and IL-4) inside the BALF of mice with asthma. A decline of OVA-dependent IgE and IgG1 inside the serum was also noticed in these mice. Rbs was proved to enhance the mRNA level of Foxp3 inside the mice lung affected with asthma while decrease that of IL-17A, IL-23, and RORγt. NF-κB pathway activation elicited by OVA was suppressed by Rbs inside the pulmonary tissues. Rbs played significantly in the reduction of airway inflammation induced by OVA which with modulating NF-κB pathway activation. PRACTICAL APPLICATIONS: Simultaneous therapy with medicine and food is strategically significant for disease prevention and treatment in traditional Chinese medicine. Rbs is a diterpene glycoside isolated from Rubus suavissimus. The anti-inflammatory and anti-asthmatic mechanism dependent of Rbs need further study clinically. The goal of current investigation is to explore the anti-inflammatory as well as anti-asthmatic activity of Rbs in mouse models of OVA-induced experimental allergic asthma. Results of the present study are scientifically supportive for the use of Rbs as an adjunctive reagent for clinical treatment of allergic asthma.
Collapse
Affiliation(s)
- Chengqiang Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, P.R. China
| | - Jia Tang
- Institution of Documentation of Chinese Traditional Medicine Research, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Bo Qian
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, P.R. China
| | - Zhen Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, P.R. China
| | - Yang Gao
- Department of Pharmacy, Northern Jiangsu People's Hospital, Yangzhou, P.R. China
| | - Jia-Le Song
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, P.R. China.,Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
25
|
Li L, Jiang M, Li Y, Su J, Li L, Qu X, Fan L. 1H-NMR Metabolomics Analysis of the Effect of Rubusoside on Serum Metabolites of Golden Hamsters on a High-Fat Diet. Molecules 2020; 25:molecules25061274. [PMID: 32168894 PMCID: PMC7143983 DOI: 10.3390/molecules25061274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 01/30/2023] Open
Abstract
Rubusoside is a natural sweetener and the active component of Rubus suavissimus. The preventive and therapeutic effect of rubusoside on high-fat diet-induced (HFD) serum metabolite changes in golden hamsters was analyzed by 1H-NMR metabolomics to explore the underlying mechanism of lipid metabolism regulation. 1H-NMR serum metabolomics analyses revealed a disturbed amino acid-, sugar-, fat-, and energy metabolism in HFD animals. Animals supplemented with rubusoside can partly reverse the metabolism disorders induced by high-fat diet and exerted good anti-hypertriglyceridemia effect by intervening in some major metabolic pathways, involving amino acid metabolism, synthesis of ketone bodies, as well as choline and 4-hydroxyphenylacetate metabolism. This study indicates that rubusoside can interfere with and normalize high-fat diet-induced metabolic changes in serum and could provide a theoretical basis to establish rubusoside as a potentially therapeutic tool able to revert or prevent lipid metabolism disorders.
Collapse
Affiliation(s)
- Li Li
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530001, China; (L.L.); (M.J.); (Y.L.); (L.L.)
| | - Manjing Jiang
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530001, China; (L.L.); (M.J.); (Y.L.); (L.L.)
| | - Yaohua Li
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530001, China; (L.L.); (M.J.); (Y.L.); (L.L.)
| | - Jian Su
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530001, China;
| | - Li Li
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530001, China; (L.L.); (M.J.); (Y.L.); (L.L.)
| | - Xiaosheng Qu
- National Engineering Laboratory of Southwest Endangered Medicinal Resources Development, Guangxi Botanical Garden of Medicinal Plants, Nanning 530023, China
- Correspondence: (X.Q.); (L.F.); Tel./Fax: +86-771-560-1290 (X.Q.); +86-771-495-3513 (L.F.)
| | - Lanlan Fan
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530001, China; (L.L.); (M.J.); (Y.L.); (L.L.)
- Correspondence: (X.Q.); (L.F.); Tel./Fax: +86-771-560-1290 (X.Q.); +86-771-495-3513 (L.F.)
| |
Collapse
|
26
|
Merino B, Fernández-Díaz CM, Cózar-Castellano I, Perdomo G. Intestinal Fructose and Glucose Metabolism in Health and Disease. Nutrients 2019; 12:E94. [PMID: 31905727 PMCID: PMC7019254 DOI: 10.3390/nu12010094] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023] Open
Abstract
The worldwide epidemics of obesity and diabetes have been linked to increased sugar consumption in humans. Here, we review fructose and glucose metabolism, as well as potential molecular mechanisms by which excessive sugar consumption is associated to metabolic diseases and insulin resistance in humans. To this end, we focus on understanding molecular and cellular mechanisms of fructose and glucose transport and sensing in the intestine, the intracellular signaling effects of dietary sugar metabolism, and its impact on glucose homeostasis in health and disease. Finally, the peripheral and central effects of dietary sugars on the gut-brain axis will be reviewed.
Collapse
Affiliation(s)
- Beatriz Merino
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
| | - Cristina M. Fernández-Díaz
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
| | - Irene Cózar-Castellano
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid 28029, Spain
| | - German Perdomo
- Instituto de Biología y Genética Molecular-IBGM (CSIC-Universidad de Valladolid), Valladolid 47003, Spain; (B.M.); (C.M.F.-D.); (G.P.)
- Departamento de Ciencias de la Salud, Universidad de Burgos, Burgos 09001, Spain
| |
Collapse
|
27
|
Goldman A, Khiste S, Freinkman E, Dhawan A, Majumder B, Mondal J, Pinkerton AB, Eton E, Medhi R, Chandrasekar V, Rahman MM, Ichimura T, Gopinath KS, Majumder P, Kohandel M, Sengupta S. Targeting tumor phenotypic plasticity and metabolic remodeling in adaptive cross-drug tolerance. Sci Signal 2019; 12:12/595/eaas8779. [PMID: 31431543 DOI: 10.1126/scisignal.aas8779] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Metastable phenotypic state transitions in cancer cells can lead to the development of transient adaptive resistance or tolerance to chemotherapy. Here, we report that the acquisition of a phenotype marked by increased abundance of CD44 (CD44Hi) by breast cancer cells as a tolerance response to routinely used cytotoxic drugs, such as taxanes, activated a metabolic switch that conferred tolerance against unrelated standard-of-care chemotherapeutic agents, such as anthracyclines. We characterized the sequence of molecular events that connected the induced CD44Hi phenotype to increased activity of both the glycolytic and oxidative pathways and glucose flux through the pentose phosphate pathway (PPP). When given in a specific order, a combination of taxanes, anthracyclines, and inhibitors of glucose-6-phosphate dehydrogenase (G6PD), an enzyme involved in glucose metabolism, improved survival in mouse models of breast cancer. The same sequence of the three-drug combination reduced the viability of patient breast tumor samples in an explant system. Our findings highlight a convergence between phenotypic and metabolic state transitions that confers a survival advantage to cancer cells against clinically used drug combinations. Pharmacologically targeting this convergence could overcome cross-drug tolerance and could emerge as a new paradigm in the treatment of cancer.
Collapse
Affiliation(s)
- Aaron Goldman
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA. .,Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.,Mitra Biotech, Integrative Immuno-Oncology Center, Woburn, MA 01801, USA
| | - Sachin Khiste
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.,Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elizaveta Freinkman
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Andrew Dhawan
- School of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Biswanath Majumder
- Mitra Biotech, Integrative Immuno-Oncology Center, Woburn, MA 01801, USA.,Mitra Biotech, 7, Service Road, Pragathi Nagar, Electronic City, Bengaluru, Karnataka 560100, India
| | - Jayanta Mondal
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.,Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Elliot Eton
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ragini Medhi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vineethkrishna Chandrasekar
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - M Mamunur Rahman
- Medical and Biological Laboratories International, Woburn, MA 01801, USA
| | - Takaharu Ichimura
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kodaganur S Gopinath
- Department of Surgical Oncology, HCG Bangalore Institute of Oncology Specialty Center, Bengaluru, Karnataka 560027, India
| | - Pradip Majumder
- Mitra Biotech, Integrative Immuno-Oncology Center, Woburn, MA 01801, USA
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Shiladitya Sengupta
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA. .,Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,Dana Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
28
|
Sugar Modification Enhances Cytotoxic Activity of PAMAM-Doxorubicin Conjugate in Glucose-Deprived MCF-7 Cells - Possible Role of GLUT1 Transporter. Pharm Res 2019; 36:140. [PMID: 31367876 PMCID: PMC6669199 DOI: 10.1007/s11095-019-2673-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/16/2019] [Indexed: 02/03/2023]
Abstract
Purpose In order to overcome the obstacles and side effects of classical chemotherapy, numerous studies have been performed to develop the treatment based on targeted transport of active compounds directly to the site of action. Since tumor cells are featured with intensified glucose metabolism, we set out to develop innovative, glucose-modified PAMAM dendrimer for the delivery of doxorubicin to breast cancer cells. Methods PAMAM-dox-glc conjugate was synthesized and characterized by 1H NMR, FT-IR, size and zeta potential measurements. The drug release rate from conjugate was evaluated by dialysis under different pH conditions. The expression level of GLUT family receptors in cells cultured in full and glucose-deprived medium was evaluated by quantitative real-time RT-PCR and flow cytometry. The cytotoxicity of conjugate in presence or absence of GLUT1 inhibitors was determined by MTT assay. Results We showed that PAMAM-dox-glc conjugate exhibits pH-dependent drug release and increased cytotoxic activity compared to free drug in cells cultured in medium without glucose. Further, we proved that these cells overexpress transporters of GLUT family. The toxic effect of conjugate was eliminated by the application of specific GLUT1 inhibitors. Conclusion Our findings revealed that the glucose moiety plays a crucial role in the recognition of cells with high expression of GLUT receptors. By selectively blocking GLUT1 transporter we showed its importance for the cytotoxic activity of PAMAM-dox-glc conjugate. These results suggest that PAMAM-glucose formulations may constitute an efficient platform for the specific delivery of anticancer drugs to tumor cells overexpressing transporters of GLUT family. Electronic supplementary material The online version of this article (10.1007/s11095-019-2673-9) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Nguyen TTH, Seo C, Kwak SH, Kim J, Kang HK, Kim SB, Kim D. Enzymatic Production of Steviol Glucosides Using β-Glucosidase and Their Applications. ENZYMES IN FOOD BIOTECHNOLOGY 2019. [PMCID: PMC7149536 DOI: 10.1016/b978-0-12-813280-7.00023-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Sweet leaf, Stevia rebaudiana Bertoni, is a perennial plant species widely known for its sweet-tastingent-kaurene type diterpenoid glycosides (steviol glucosides). Steviol glucosides include rubusoside (Ru), stevioside (Ste), and rebaudioside (Reb), which have immunomodulatory capability and protective effects against hyperglycemia, hypertension, inflammation, tumors, and diarrhea. In addition, they can enhance the solubility of epotoside, liquiritin, paclitaxel, curcuminoids, quercetin, and wheat bran flavonoids, thus increasing their permeability. The hydrolysis of three glucosyl groups at positions C13 and C19 of Ste will produce steviolbioside, steviol, isosteviol, steviol mono-glucoside, or Ru. S. rebaudiana contains these hydrolyzed products in trace amounts. This chapter describes recent advances in the preparation of various steviol glycosides from Ste by using different β-glycosidases, with particular focus on their potential industrial applications as natural solubilizers of insoluble compounds. Furthermore, the reaction mechanism of β-glycosidases and their kinetic properties are summarized.
Collapse
Affiliation(s)
- Thi Thanh Hanh Nguyen
- The Institute of Food Industrialization, Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Changseop Seo
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - So-Hyung Kwak
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Jeesoo Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea
| | - Hee-Kyoung Kang
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Seong-Bo Kim
- CJ CheilJedang, Life Ingredient and Material Research Institute, Suwon, South Korea
| | - Doman Kim
- The Institute of Food Industrialization, Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang-gun, South Korea,Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang-gun, South Korea
| |
Collapse
|
30
|
Zheng H, Wu J, Huang H, Meng C, Li W, Wei T, Su Z. Metabolomics analysis of the protective effect of rubusoside on palmitic acid-induced lipotoxicity in INS-1 cells using UPLC-Q/TOF MS. Mol Omics 2019; 15:222-232. [DOI: 10.1039/c9mo00029a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetes is one of the most severe chronic diseases worldwide.
Collapse
Affiliation(s)
- Hua Zheng
- Life Sciences Institute
- Guangxi Medical University
- Nanning 530021
- China
| | - Jinxia Wu
- Pharmaceutical College
- Guangxi Medical University
- Nanning 530021
- China
| | - Hong Huang
- The First Affiliated Hospital
- Guangxi Medical University
- Nanning 530021
- China
| | - Chunmei Meng
- Life Sciences Institute
- Guangxi Medical University
- Nanning 530021
- China
| | - Weidong Li
- Life Sciences Institute
- Guangxi Medical University
- Nanning 530021
- China
| | - Tianli Wei
- Pharmaceutical College
- Guangxi Medical University
- Nanning 530021
- China
| | - Zhiheng Su
- Pharmaceutical College
- Guangxi Medical University
- Nanning 530021
- China
| |
Collapse
|
31
|
Intestinal Saturated Long-Chain Fatty Acid, Glucose and Fructose Transporters and Their Inhibition by Natural Plant Extracts in Caco-2 Cells. Molecules 2018; 23:molecules23102544. [PMID: 30301205 PMCID: PMC6222386 DOI: 10.3390/molecules23102544] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 09/29/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023] Open
Abstract
The intestinal absorption of fatty acids, glucose and fructose is part of the basic requirements for the provision of energy in the body. High access of saturated long-chain fatty acids (LCFA), glucose and fructose can facilitate the development of metabolic diseases, particularly the metabolic syndrome and type-2 diabetes mellitus (T2DM). Research has been done to find substances which decelerate or inhibit intestinal resorption of these specific food components. Promising targets are the inhibition of intestinal long-chain fatty acid (FATP2, FATP4), glucose (SGLT1, GLUT2) and fructose (GLUT2, GLUT5) transporters by plant extracts and by pure substances. The largest part of active components in plant extracts belongs to the group of polyphenols. This review summarizes the knowledge about binding sites of named transporters and lists the plant extracts which were tested in Caco-2 cells regarding uptake inhibition.
Collapse
|
32
|
Schmidl S, Iancu CV, Choe JY, Oreb M. Ligand Screening Systems for Human Glucose Transporters as Tools in Drug Discovery. Front Chem 2018; 6:183. [PMID: 29888221 PMCID: PMC5980966 DOI: 10.3389/fchem.2018.00183] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/07/2018] [Indexed: 12/22/2022] Open
Abstract
Hexoses are the major source of energy and carbon skeletons for biosynthetic processes in all kingdoms of life. Their cellular uptake is mediated by specialized transporters, including glucose transporters (GLUT, SLC2 gene family). Malfunction or altered expression pattern of GLUTs in humans is associated with several widespread diseases including cancer, diabetes and severe metabolic disorders. Their high relevance in the medical area makes these transporters valuable drug targets and potential biomarkers. Nevertheless, the lack of a suitable high-throughput screening system has impeded the determination of compounds that would enable specific manipulation of GLUTs so far. Availability of structural data on several GLUTs enabled in silico ligand screening, though limited by the fact that only two major conformations of the transporters can be tested. Recently, convenient high-throughput microbial and cell-free screening systems have been developed. These remarkable achievements set the foundation for further and detailed elucidation of the molecular mechanisms of glucose transport and will also lead to great progress in the discovery of GLUT effectors as therapeutic agents. In this mini-review, we focus on recent efforts to identify potential GLUT-targeting drugs, based on a combination of structural biology and different assay systems.
Collapse
Affiliation(s)
- Sina Schmidl
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Cristina V Iancu
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Jun-Yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Mislav Oreb
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
33
|
Abstract
Increased understanding of fructose metabolism, which begins with uptake via the intestine, is important because fructose now constitutes a physiologically significant portion of human diets and is associated with increased incidence of certain cancers and metabolic diseases. New insights in our knowledge of intestinal fructose absorption mediated by the facilitative glucose transporter GLUT5 in the apical membrane and by GLUT2 in the basolateral membrane are reviewed. We begin with studies related to structure as well as ligand binding, then revisit the controversial proposition that apical GLUT2 is the main mediator of intestinal fructose absorption. The review then describes how dietary fructose may be sensed by intestinal cells to affect the expression and activity of transporters and fructolytic enzymes, to interact with the transport of certain minerals and electrolytes, and to regulate portal and peripheral fructosemia and glycemia. Finally, it discusses the potential contributions of dietary fructose to gastrointestinal diseases and to the gut microbiome.
Collapse
Affiliation(s)
- Ronaldo P Ferraris
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey 07946, USA;
| | - Jun-Yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, North Chicago, Illinois 60064, USA;
| | - Chirag R Patel
- Independent Drug Safety Consulting, Wilmington, Delaware 19803, USA;
| |
Collapse
|
34
|
Synthesis and characterization of novel astragalin galactosides using β-galactosidase from Bacillus circulans. Enzyme Microb Technol 2017; 103:59-67. [DOI: 10.1016/j.enzmictec.2017.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/07/2017] [Accepted: 05/08/2017] [Indexed: 01/18/2023]
|
35
|
Abstract
Facilitative carbohydrate transporters-Gluts-have received wide attention over decades due to their essential role in nutrient uptake and links with various metabolic disorders, including diabetes, obesity, and cancer. Endeavors directed towards understanding the mechanisms of Glut-mediated nutrient uptake have resulted in a multidisciplinary research field spanning protein chemistry, chemical biology, organic synthesis, crystallography, and biomolecular modeling. Gluts became attractive targets for cancer research and medicinal chemistry, leading to the development of new approaches to cancer diagnostics and providing avenues for cancer-targeting therapeutics. In this review, the current state of knowledge of the molecular interactions behind Glut-mediated sugar uptake, Glut-targeting probes, therapeutics, and inhibitors are discussed.
Collapse
Affiliation(s)
- Marina Tanasova
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, MI, 49931, USA
| | - Joseph R Fedie
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, MI, 49931, USA
| |
Collapse
|
36
|
Tripp J, Essl C, Iancu CV, Boles E, Choe JY, Oreb M. Establishing a yeast-based screening system for discovery of human GLUT5 inhibitors and activators. Sci Rep 2017; 7:6197. [PMID: 28740135 PMCID: PMC5524692 DOI: 10.1038/s41598-017-06262-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/05/2017] [Indexed: 01/08/2023] Open
Abstract
Human GLUT5 is a fructose-specific transporter in the glucose transporter family (GLUT, SLC2 gene family). Its substrate-specificity and tissue-specific expression make it a promising target for treatment of diabetes, metabolic syndrome and cancer, but few GLUT5 inhibitors are known. To identify and characterize potential GLUT5 ligands, we developed a whole-cell system based on a yeast strain deficient in fructose uptake, in which GLUT5 transport activity is associated with cell growth in fructose-based media or assayed by fructose uptake in whole cells. The former method is convenient for high-throughput screening of potential GLUT5 inhibitors and activators, while the latter enables detailed kinetic characterization of identified GLUT5 ligands. We show that functional expression of GLUT5 in yeast requires mutations at specific positions of the transporter sequence. The mutated proteins exhibit kinetic properties similar to the wild-type transporter and are inhibited by established GLUT5 inhibitors N-[4-(methylsulfonyl)-2-nitrophenyl]-1,3-benzodioxol-5-amine (MSNBA) and (−)-epicatechin-gallate (ECG). Thus, this system has the potential to greatly accelerate the discovery of compounds that modulate the fructose transport activity of GLUT5.
Collapse
Affiliation(s)
- Joanna Tripp
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438, Frankfurt am Main, Germany
| | - Christine Essl
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438, Frankfurt am Main, Germany
| | - Cristina V Iancu
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Eckhard Boles
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438, Frankfurt am Main, Germany
| | - Jun-Yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Mislav Oreb
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
37
|
Reassessment of GLUT7 and GLUT9 as Putative Fructose and Glucose Transporters. J Membr Biol 2017; 250:171-182. [PMID: 28083649 DOI: 10.1007/s00232-016-9945-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/22/2016] [Indexed: 02/01/2023]
Abstract
Although increased dietary fructose consumption is associated with metabolic impairments, the mechanisms and regulation of intestinal fructose absorption are poorly understood. GLUT5 is considered to be the main intestinal fructose transporter. Other GLUT family members, such as GLUT7 and GLUT9 are also expressed in the intestine and were shown to transport fructose and glucose. A conserved isoleucine-containing motif (NXI) was proposed to be essential for fructose transport capacity of GLUT7 and GLUT9 but also of GLUT2 and GLUT5. In assessing whether human GLUT2, GLUT5, GLUT7, and GLUT9 are indeed fructose transporters, we expressed these proteins in Xenopus laevis oocytes. Stably transfected NIH-3T3 fibroblasts were used as second expression system. In proving the role of the NXI motif, variants p.I322V of GLUT2 and p.I296V of GLUT5 were tested as well. Sugar transport was measured by radiotracer flux assays or by metabolomics analysis of cell extracts by GC-MS. Fructose and glucose uptakes by GLUT7 were not increased in both expression systems. In search for the physiological substrate of GLUT7, cells overexpressing the protein were exposed to various metabolite mixtures, but we failed to identify a substrate. Although urate transport by GLUT9 could be shown, neither fructose nor glucose transport was detectable. Fructose uptake was decreased by the GLUT2 p.I322V variant, but remained unaffected in the p.I296V GLUT5 variant. Thus, our work does not find evidence that GLUT7 or GLUT9 transport fructose or glucose or that the isoleucine residue determines fructose specificity. Rather, the physiological substrate of GLUT7 awaits to be discovered.
Collapse
|
38
|
Labak CM, Wang PY, Arora R, Guda MR, Asuthkar S, Tsung AJ, Velpula KK. Glucose transport: meeting the metabolic demands of cancer, and applications in glioblastoma treatment. Am J Cancer Res 2016; 6:1599-608. [PMID: 27648352 PMCID: PMC5004066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 06/06/2023] Open
Abstract
GLUT1, and to a lesser extent, GLUT3, appear to be interesting targets in the treatment of glioblastoma multiforme. The current review aims to give a brief history of the scientific community's understanding of these glucose transporters and to relate their importance to the metabolic changes that occur as a result of cancer. One of the primary changes that occurs in cancer, the Warburg Effect, is characterized by an extreme shift toward glycolysis from the usual reliance on oxidative phosphorylation and is currently being investigated to target the upstream and downstream factors responsible for Warburg-induced changes. Further, it aims to explain the differential expression of GLUT1 and GLUT3 in glioblastoma tissue, and how these modulations in expression can serve as targets to restore a more normal metabolism. Additionally, hypoxia-induced factor-1α's (HIF1α) role in a number of transcriptional changes typical to GBM will be discussed, including its role in GLUT upregulation. Finally, the four known subtypes of GBM [proneural, neural, mesenchymal, and classical] will be characterized in order to discuss how metabolic changes differ in each subtype. These changes have the potential to be selectively targeted in order to provide specificity to the clinical treatment options in GBM.
Collapse
Affiliation(s)
- Collin M Labak
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Paul Y Wang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Rishab Arora
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Maheedhara R Guda
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Andrew J Tsung
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Illinois Neurological InstitutePeoria, IL, USA
| | - Kiran K Velpula
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Microbiology, Yogi Vemana UniversityKadapa, India
| |
Collapse
|
39
|
George Thompson AM, Ursu O, Babkin P, Iancu CV, Whang A, Oprea TI, Choe JY. Discovery of a specific inhibitor of human GLUT5 by virtual screening and in vitro transport evaluation. Sci Rep 2016; 6:24240. [PMID: 27074918 PMCID: PMC4831007 DOI: 10.1038/srep24240] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/23/2016] [Indexed: 02/07/2023] Open
Abstract
GLUT5, a fructose-transporting member of the facilitative glucose transporter (GLUT, SLC2) family, is a therapeutic target for diabetes and cancer but has no potent inhibitors. We virtually screened a library of 6 million chemicals onto a GLUT5 model and identified N-[4-(methylsulfonyl)-2-nitrophenyl]-1,3-benzodioxol-5-amine (MSNBA) as an inhibitor of GLUT5 fructose transport in proteoliposomes. MSNBA inhibition was specific to GLUT5; this inhibitor did not affect the fructose transport of human GLUT2 or the glucose transport of human GLUT1-4 or bacterial GlcPSe. In MCF7 cells, a human breast cancer cell line, MSNBA competitively inhibited GLUT5 fructose uptake with a KI of 3.2 ± 0.4 μM. Ligand docking, mutagenesis and functional studies indicate that MSNBA binds near the active site and inhibitor discrimination involves H387 of GLUT5. Thus, MSNBA is a selective and potent inhibitor of fructose transport via GLUT5, and the first chemical probe for this transporter. Our data indicate that active site differences in GLUT members could be exploited to further enhance ligand specificity.
Collapse
Affiliation(s)
- Alayna M George Thompson
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Oleg Ursu
- Department of Internal Medicine, Translational Informatics Division, MSC09 5025, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Petr Babkin
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Cristina V Iancu
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Alex Whang
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Tudor I Oprea
- Department of Internal Medicine, Translational Informatics Division, MSC09 5025, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Jun-yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| |
Collapse
|
40
|
Jordan P, Choe JY, Boles E, Oreb M. Hxt13, Hxt15, Hxt16 and Hxt17 from Saccharomyces cerevisiae represent a novel type of polyol transporters. Sci Rep 2016; 6:23502. [PMID: 26996892 PMCID: PMC4800717 DOI: 10.1038/srep23502] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/09/2016] [Indexed: 12/03/2022] Open
Abstract
The genome of S. cerevisae encodes at least twenty hexose transporter-like proteins. Despite extensive research, the functions of Hxt8-Hxt17 have remained poorly defined. Here, we show that Hxt13, Hxt15, Hxt16 and Hxt17 transport two major hexitols in nature, mannitol and sorbitol, with moderate affinities, by a facilitative mechanism. Moreover, Hxt11 and Hxt15 are capable of transporting xylitol, a five-carbon polyol derived from xylose, the most abundant pentose in lignocellulosic biomass. Hxt11, Hxt13, Hxt15, Hxt16 and Hxt17 are phylogenetically and functionally distinct from known polyol transporters. Based on docking of polyols to homology models of transporters, we propose the architecture of their active site. In addition, we determined the kinetic parameters of mannitol and sorbitol dehydrogenases encoded in the yeast genome, showing that they discriminate between mannitol and sorbitol to a much higher degree than the transporters.
Collapse
Affiliation(s)
- Paulina Jordan
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438 Frankfurt am Main, Germany
| | - Jun-Yong Choe
- Department of Biochemistry and Molecular Biology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Eckhard Boles
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438 Frankfurt am Main, Germany
| | - Mislav Oreb
- Institute of Molecular Biosciences, Goethe University, Max-von-Laue Straße 9, 60438 Frankfurt am Main, Germany
| |
Collapse
|