1
|
Peng Y, Lin C, Zhang B, Yan L, Zhang B, Zhao C, Qiu L. Characteristics and preliminary immune function of SRA5 in Lateolabrax maculatus. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110266. [PMID: 40064212 DOI: 10.1016/j.fsi.2025.110266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 03/26/2025]
Abstract
Scavenger receptors (SRs) are crucial for pattern recognition in the innate immune system. However, the role of Scavenger Receptors class A member 5 (SRA5) in the immunological response of bony fish to pathogen invasion remains unclear. This study identified and characterized the SRA5 of Lateolabrax maculatus (LmSRA5) from its transcriptome database. LmSRA5 has a 1494 bp open reading frame, encodes 497 amino acids, has a molecular weight of 55.01 kDa, and contains a collagen domain and a conserved Scavenger Receptor Cysteine-Rich domain. LmSRA5 exhibited high sequence similarity to previously reported SRA5 genes. LmSRA5 exhibited high sequence similarity to previously reported SRA5 genes. LmSRA5 is primarily localized in the cytoplasm, with its encoded proteins distributed in both the cytoplasm and the cell membrane. LmSRA5 was expressed in all tissues. The highest expression was observed in the pituitary gland, with significant levels in the stomach, intestines, liver, and kidney. LmSRA5 expression in the head kidney, spleen, blood, and intestines initially increased, then decreased following infection with Aeromonas veronii. The binding affinity of LmSRA5 for A. veronii was enhanced by increasing concentrations of the extracellular domain recombinant LmSRA5. Knockdown and overexpression experiments in liver cells demonstrated that LmSRA5 significantly regulates the expression of IL-8 and c-Jun. LmSRA5 participates in the immune response by recognizing pathogen-associated molecular patterns (PAMPs) and contributes to immune regulation through modulation IL-8 and c-Jun. This study offers valuable insights into the role of SRA5 in pathogen resistance and immune regulation in bony fish, thereby contributing to the advancement of aquaculture under escalating disease pressures.
Collapse
Affiliation(s)
- Yangtao Peng
- College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China
| | - Changhong Lin
- College of Aqua-life Science and Technology, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China.
| | - Lulu Yan
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China.
| | - Chao Zhao
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China
| | - Lihua Qiu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, PR China; Sanya Tropical Fisheries Research Institute, Sanya, PR China; Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, Chinese Academy of Fishery Science, Beijing, PR China.
| |
Collapse
|
2
|
Zhang T, Zhang J, Wang H, Zou T, Cheng S, Yu Y, Wu J, Pan Y, Duan X, Wang H. Lateral Organization and Dynamics of the Realistic Plasma Membrane. J Phys Chem B 2025. [PMID: 40397703 DOI: 10.1021/acs.jpcb.5c00909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Large-scale simulations of realistic crowded cell membranes can bridge the gap between the simulations and experiments. However, the compositional complexity and structural asymmetry of cell membranes continue to pose significant challenges in computational biology. Recent advances in understanding native membranes, including their composition and protein structures, enable us to construct a highly realistic model of the mammalian plasma membrane. Using this model, we explore the organization and dynamics of biological cell membranes at the molecular level. We found that the interaction preferences of protein-lipid mediate the formation of dynamic clusters of nonrandomly distributed proteins, accompanied by heterogeneous structural properties and anomalous diffusion. These evolving dynamic clusters intertwine to form a highly complex and continuously changing protein network. Our study provides significant insights into the intricate lateral dynamic organization of cell membranes.
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Jinrui Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
| | - Huili Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
| | - Tianyi Zou
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Sihang Cheng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Yang Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Junbo Wu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Yangang Pan
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Xiaozheng Duan
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| |
Collapse
|
3
|
Andersen CG, Bavnhøj L, Brag S, Bohush A, Chrenková A, Driller JH, Pedersen BP. Comparative analysis of STP6 and STP10 unravels molecular selectivity in sugar transport proteins. Proc Natl Acad Sci U S A 2025; 122:e2417370122. [PMID: 40279393 PMCID: PMC12054785 DOI: 10.1073/pnas.2417370122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 03/20/2025] [Indexed: 04/27/2025] Open
Abstract
The distribution of sugars is crucial for plant energy, signaling, and defense mechanisms. Sugar Transport Proteins (STPs) are Sugar Porters (SPs) that mediate proton-driven cellular uptake of glucose. Some STPs also transport fructose, while others remain highly selective for only glucose. What determines this selectivity, allowing STPs to distinguish between compounds with highly similar chemical composition, remains unknown. Here, we present the structure of Arabidopsis thaliana STP6 in an inward-occluded conformational state with glucose bound and demonstrate its role as both a glucose and fructose transporter. We perform a comparative analysis of STP6 with the glucose-selective STP10 using in vivo and in vitro systems, demonstrating how different experimental setups strongly influence kinetic transport properties. We analyze the properties of the monosaccharide binding site and show that the position of a single methyl group in the binding site is sufficient to shuffle glucose and fructose specificity, providing detailed insights into the fine-tuned dynamics of affinity-induced specificity for sugar uptake. Altogether, these findings enhance our understanding of sugar selectivity in STPs and more broadly SP proteins.
Collapse
Affiliation(s)
| | - Laust Bavnhøj
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus CDK-8000, Denmark
| | - Søren Brag
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus CDK-8000, Denmark
| | - Anastasiia Bohush
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus CDK-8000, Denmark
| | - Adriana Chrenková
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus CDK-8000, Denmark
| | - Jan Heiner Driller
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus CDK-8000, Denmark
| | | |
Collapse
|
4
|
Sinclair LV, Cantrell DA. Protein Synthesis and Metabolism in T Cells. Annu Rev Immunol 2025; 43:343-366. [PMID: 40279310 DOI: 10.1146/annurev-immunol-082323-035253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
T lymphocytes are essential for immune responses to pathogens and tumors. Their ability to rapidly clonally expand and differentiate to effector cells following infection, and then to curb effector function following infection clearance, is fundamental for adaptive immunity. Proteome remodeling in response to immune activation is a fundamental mechanism that allows T cells to swiftly reprogram for acquisition of effector function and is possible only because antigen receptor- and cytokine-driven signal transduction pathways can trigger massive increases in protein synthesis. Equally, the ability to repress protein synthesis supports a return to quiescence once pathogens are cleared to avoid autoimmunity and to generate memory T cell populations. This review discusses what is known about T cell proteomes and the regulatory mechanisms that control protein synthesis in T cells. The focus is on how this fundamental process is dynamically controlled to ensure immune homeostasis.
Collapse
Affiliation(s)
- Linda V Sinclair
- Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom;
| | - Doreen A Cantrell
- Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom;
| |
Collapse
|
5
|
Ruan Z, Wang Y, Shi L, Yang XJ. Progress of research on glucose transporter proteins in hepatocellular carcinoma. World J Hepatol 2025; 17:104715. [PMID: 40177207 PMCID: PMC11959659 DOI: 10.4254/wjh.v17.i3.104715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumour with high prevalence and mortality rate worldwide. Metabolic reprogramming of cancer cells may be a major factor in the process of this disease. Glucose transporter proteins (GLUTs) are members of the major facilitator superfamily of membrane transporters, playing a pivotal role in the metabolic reprogramming and tumour progression in HCC. This review discusses the advances in the study of GLUTs in HCC, including the expression patterns, functions and possibilities of GLUTs. In HCC, the expression levels of GLUTs are closely associated with tumour aggressiveness, metabolic reprogramming and prognosis. A series of inhibitors have been demonstrated efficacy in inhibiting HCC cell growth and glucose uptake in in vitro and in vivo models. These inhibitors offer a novel approach to HCC treatment by reducing the glucose metabolism of tumour cells, thereby impeding tumour growth, and concurrently enhancing the sensitivity to chemotherapeutic agents. This reminds us of the urgent need to elucidate GLUTs' roles in HCC and to determine the most effective ways to translate these findings into clinical practice.
Collapse
Affiliation(s)
- Zheng Ruan
- The First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yan Wang
- Division of Personnel, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu Province, China
| | - Lei Shi
- Department of General Surgery, The Second people's Hospital of Lanzhou, Lanzhou 730000, Gansu Province, China
| | - Xiao-Jun Yang
- Department of General Surgery, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
6
|
Van Goor A, Pasternak A, Walker KE, Chick S, Harding JCS, Lunney JK. Altered structural and transporter-related gene expression patterns in the placenta play a role in fetal demise during Porcine reproductive and respiratory syndrome virus infection. BMC Genomics 2025; 26:279. [PMID: 40119254 PMCID: PMC11927291 DOI: 10.1186/s12864-025-11397-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/21/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Porcine reproductive and respiratory syndrome virus (PRRSV) can be transmitted across the maternal-fetal-interface from an infected gilt to her fetuses. Although fetal infection status and disease outcomes vary, the mechanisms are not completely understood. The objective was to assess targeted placental structural and transporter-related gene expression patterns. At day 85 of gestation pregnant pigs were challenged with PRRSV, and at 12 days post maternal infection sows and fetuses were sacrificed, and the placental tissue was collected. Grouping of fetuses was by preservation status and PRRS viral load (VL): control (CTRL, n = 14), viable and low VL fetus (VIA_LVF, n = 15), viable and high VL fetus (VIA_HVF, n = 21), meconium mild and low VL fetus (MECm_LVF, n = 14), meconium mild and high VL fetus (MECm_HVF, n = 14), and meconium severe and high VL fetus (MECs_HVF, n = 13). NanoString was used to evaluate the expression of 86 genes: actin cytoskeleton signaling, arachidonic acid pathway, integrin signaling, intercellular junctions, transporters, and VEGF signaling. Statistical analyses were performed using Limma with P ≤ 0.05 considered significant. RESULTS We identified 1, 7, 0, 29, and 39 differentially expressed genes in VIA_LVF, VIA_HVF, MECm_LVF, MECm_HVF, and MECs_HVF, respectively, contrasted to CTRL. Placental transporter genes were significantly impacted (i.e., downregulation of SLC1A3, SLC1A5, SLC2A1, SLC2A3, SLC2A5, SLC2A10, SLC2A12, SLC7A4, SLC16A5, SLC16A10, and SLC27A6; and upregulation of SLC2A2, SLC16A3, and SLC27A4), compared to CTRL. Actin cytoskeleton signaling (ARHGEF6 and ARHGEF7), arachidonic acid (PTGES3 and PTGIS), integrin signaling (FN1 and ITGB6), intercellular junctions (CDH3 and CDH11), and VEGF signaling (MAPK3 and HPSE) gene groupings were significantly impacted, compared to CTRL. CONCLUSION Data reported here indicate that fetal PRRSV infection levels rather than fetal demise is necessary for transcriptional dysregulation of the fetal placenta, with a tendency towards more downregulation in the target gene sets among susceptible fetuses. These results generally support that in susceptible fetuses there is altered solute transportation, placental structural integrity, and reduced angiogenesis. The data described here is associated with fetal PRRS resistance/resilience and susceptibility.
Collapse
Affiliation(s)
- Angelica Van Goor
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, ARS, USDA, Beltsville, MD, USA
- Division of Animal Systems, Institute of Food Production and Sustainability, NIFA, USDA, Kansas City, MO, USA
| | - Alex Pasternak
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Kristen E Walker
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, ARS, USDA, Beltsville, MD, USA
| | - Shannon Chick
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, ARS, USDA, Beltsville, MD, USA
| | - John C S Harding
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Joan K Lunney
- Animal Parasitic Diseases Laboratory, Beltsville Agricultural Research Center, ARS, USDA, Beltsville, MD, USA.
| |
Collapse
|
7
|
Quan C, Jiang X. The molecular mechanism underlying the human glucose facilitators inhibition. VITAMINS AND HORMONES 2025; 128:49-92. [PMID: 40097253 DOI: 10.1016/bs.vh.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Glucose is the primary energy substrate and an essential precursor for cellular metabolism. Maintaining glucose homeostasis necessitates the presence of glucose transporters, as the hydrophilic nature of glucose prevents its passage across the cell membrane. The GLUT family is a crucial group of glucose transporters that facilitate glucose diffusion along the transmembrane glucose concentration gradient. Dysfunction in GLUTs is associated with diseases, such as GLUT1 deficiency syndrome, Fanconi-Bickel syndrome, and type 2 diabetes. Furthermore, elevated expression of GLUTs fuels aerobic glycolysis, known as the Warburg effect, in various types of cancers, making GLUT isoforms possible targets for antineoplastic therapies. To date, 30 GLUT and homolog structures have been released on the Protein Data Bank (PDB), showcasing multiple conformational and ligand-binding states. These structures elucidate the molecular mechanisms underlying substrate recognition, the alternating access cycle, and transport inhibition. Here, we summarize the current knowledge of human GLUTs and their role in cancer, highlighting recent advances in the structural characterization of GLUTs. We also compare the inhibition mechanisms of exofacial and endofacial GLUT inhibitors, providing insights into the design and optimization of GLUT inhibitors for therapeutic applications.
Collapse
Affiliation(s)
- Cantao Quan
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, The Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, P.R. China
| | - Xin Jiang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, The Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, P.R. China.
| |
Collapse
|
8
|
Khandelwal NK, Gupta M, Kumar P, Balasubramani SG, Echeverria I, Stroud RM. Structural basis of disease mutation and substrate recognition by the human SLC2A9 transporter. Proc Natl Acad Sci U S A 2025; 122:e2418282122. [PMID: 39937868 PMCID: PMC11848319 DOI: 10.1073/pnas.2418282122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/10/2024] [Indexed: 02/14/2025] Open
Abstract
Urate provides ~50% of the reducing potential in human and primate plasma which is key to detoxifying reactive oxygen by-products of cellular metabolism. Urate is the endpoint of purine metabolism in primates, and its concentration in plasma is a balance between excretion from kidney and intestine, and subsequent reabsorption in and through cells of kidney proximal tubules to maintain a regulated concentration in plasma. SLC2A9 is the primary transporter that returns urate from the basolateral side of kidney tubule cells back to plasma. A shorter splice variant of SLC2A9 is directed to the apical surface where several transporters recapture urate from the tubule back into cells. Too high a concentration in plasma causes hyperuricemia, is linked to gout, and favors kidney stone formation. To understand the molecular basis of uric acid transport and the role of disease-causing mutations in SLC2A9, we determined structures of human SLC2A9 in its apo form, and its urate-bound form by cryo-EM, at resolution of 3.3 Å and 4.1 Å respectively. Both structures are captured in an inward open conformation. Using the inward-facing structure as a template we modeled the outward-facing conformation to understand the alternating access mechanism. Alternative salt bridge pairs on the cytoplasmic side suggest a mechanism that can balance the energetics of the inward open and outward open states. The location of disease-causing mutants suggests their role in impacting function. Our structures elucidate the molecular basis for urate selectivity and transport and provide a platform for future structure-based drug discovery aimed at reducing plasma urate levels in diseases of hyperuricemia and gout.
Collapse
Affiliation(s)
- Nitesh Kumar Khandelwal
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA94143
| | - Meghna Gupta
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA94143
| | - Paras Kumar
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA94143
| | - Sree Ganesh Balasubramani
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA94158
| | - Ignacia Echeverria
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA94158
| | - Robert M. Stroud
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA94143
| |
Collapse
|
9
|
Elbahnsi A, Dudas B, Callebaut I, Hinzpeter A, Miteva MA. ATP-Binding Cassette and Solute Carrier Transporters: Understanding Their Mechanisms and Drug Modulation Through Structural and Modeling Approaches. Pharmaceuticals (Basel) 2024; 17:1602. [PMID: 39770445 PMCID: PMC11676857 DOI: 10.3390/ph17121602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
The ATP-binding cassette (ABC) and solute carrier (SLC) transporters play pivotal roles in cellular transport mechanisms, influencing a wide range of physiological processes and impacting various medical conditions. Recent advancements in structural biology and computational modeling have provided significant insights into their function and regulation. This review provides an overview of the current knowledge of human ABC and SLC transporters, emphasizing their structural and functional relationships, transport mechanisms, and the contribution of computational approaches to their understanding. Current challenges and promising future research and methodological directions are also discussed.
Collapse
Affiliation(s)
- Ahmad Elbahnsi
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| | - Balint Dudas
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| | - Isabelle Callebaut
- Muséum National d’Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie—IMPMC, Sorbonne Université, 75005 Paris, France
| | - Alexandre Hinzpeter
- CNRS, INSERM, Institut Necker Enfants Malades—INEM, Université Paris Cité, 75015 Paris, France
| | - Maria A. Miteva
- Inserm U1268 MCTR, CiTCoM UMR 8038 CNRS, Université Paris Cité, 75006 Paris, France
| |
Collapse
|
10
|
Zhang D, Guo J, Lin Z, Yan H, Peng K, Fei L, Zhai Q, Zou D, Zhong J, Ding Y, Ye H, Wang P, Wang J, Luo S, Li B, Li B, Liao W. SLC2A1 variants cause late-onset epilepsy and the genetic-dependent stage feature : For the China Epilepsy Gene 1.0 Project. ACTA EPILEPTOLOGICA 2024; 6:38. [PMID: 40217381 PMCID: PMC11960290 DOI: 10.1186/s42494-024-00177-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/02/2024] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND The SLC2A1 gene plays a vital role in brain energy metabolism. SLC2A1 variants have been reported to be associated with early-onset refractory seizures. This study aims to explore the association between the SLC2A1 gene and late-onset epilepsy. METHODS Trios-based whole-exome sequencing was performed on patients with epilepsy without acquired etiologies. The pathogenicity of the variants was assessed according to the American College of Medical Genetics and Genomics (ACMG) guidelines. RESULTS A total of 14 heterozygous SLC2A1 variants were identified in 16 unrelated families. The variants were evaluated as "pathogenic" or "likely pathogenic" according to the ACMG guidelines. Ten cases (62.5%) presented with infantile onset seizures and developmental delay/intellectual disability and were diagnosed with developmental and epileptic encephalopathy (DEE). The other six cases (37.5%) exhibited late-onset seizures and normal development. They were diagnosed with idiopathic partial epilepsy (n = 2) or idiopathic generalized epilepsy (n = 4). Further analysis showed that DEE-associated variants tended to cluster in the transmembrane region, whereas the mild epilepsy-associated variants tended to locate in regions outside the transmembrane region, suggesting a potential molecular sub-regional effect. A total of 15 cases had delayed diagnosis, with the longest delay being 22 years. The SLC2A1 expression stage, which is expressed at relatively high level throughout the whole life span, from the embryonic to adult stages with two peaks at approximately four and 14 years, is generally consistent with the seizure onset age. In addition, patients with early-onset age had variants that were potentially associated with severe damage, suggesting a potential correlation between the age of disease onset and the damaging effects of the variants. CONCLUSIONS SLC2A1 variants are associated with late-onset epilepsy, which is consistent with the genetic-dependent stage feature of SLC2A1. Early genetic diagnosis is important for treatment of patients with SLC2A1 variants.
Collapse
Affiliation(s)
- Dongming Zhang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Jing Guo
- Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou, 510520, China
| | - Zisheng Lin
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Hongjun Yan
- Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou, 510520, China
| | - Kai Peng
- Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou, 510520, China
| | - Linxia Fei
- Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou, 510520, China
| | - Qiongxiang Zhai
- Department of Pediatrics, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Dongfang Zou
- Epilepsy Center and Department of Neurology, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Jiayi Zhong
- Department of Neuroelectrophysiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yan Ding
- Department of Neurology, The First Affiliated Hospital of Jinan University, Clinical Neuroscience Institute of Jinan University, Guangzhou, 510630, China
| | - Hong Ye
- Epilepsy Center of Foshan First Hospital, Foshan, 528000, China
| | - Pengyu Wang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Jie Wang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Sheng Luo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Bingmei Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Bin Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China.
| | - Weiping Liao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
11
|
Mardones L. Transport of dehydroascorbic acid by glucose transporters GLUTs. VITAMINS AND HORMONES 2024; 128:155-180. [PMID: 40097249 DOI: 10.1016/bs.vh.2024.09.002] [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
Vitamin C is a crucial water-soluble antioxidant and an essential cofactor for enzymes like proline and lysine hydroxylases, playing a vital role in cellular physiology. While sodium-dependent ascorbate co-transporters (SVCT1 and SVCT2) are pivotal for vitamin C absorption and bioavailability, dehydroascorbic acid transporters within the facilitative glucose transporter (GLUT) family complement these functions and are relevant in various cellular, tissue-specific, or pathological contexts. This review focuses on comparing the structural and functional characteristics of GLUTs involved in glucose, dehydroascorbic acid and other substrate transport. It also presents evidence of the physiological and pathophysiological roles of dehydroascorbic acid transporters. Improved understanding of these transporters has the potential to advance strategies for preventing, diagnosing, and treating prevalent diseases such as cancer.
Collapse
Affiliation(s)
- L Mardones
- Biomedical Sciences Research Laboratory, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile Research Centre of Biodiversity and Sustainable Environment (CIBAS). Universidad Católica de la Santísima Concepción, Concepción, Chile.
| |
Collapse
|
12
|
Ferrari A, Filoni J, Di Dedda C, Piemonti L, Monti P. Ketone bodies rescue T cell impairments induced by low glucose availability. Eur J Nutr 2024; 63:2815-2825. [PMID: 39105784 DOI: 10.1007/s00394-024-03469-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 07/14/2024] [Indexed: 08/07/2024]
Abstract
PURPOSE Ketogenic diets are proposed as a therapeutic approach for type 1 and type 2 diabetes due to their low glucose intake. However, their potential effects on the immune system need investigation. This study aims to explore how glucose concentration and beta-hydroxybutyrate (BHB) impact T cell phenotype, metabolism, and function, with a focus on systemic inflammatory response (T2D) and autoimmunity (T1D). METHODS T cells from healthy donors were cultured in vitro under varying glucose concentrations with or without BHB. Flow cytometry was employed to analyze changes in T cell phenotype, while proliferation was evaluated through a CFSE dilution assay. Additionally, we used a novel flow cytometry method allowing a direct assessment of T cell metabolism. RESULTS Culturing T cells in low glucose concentrations revealed their dependency on glucose metabolism, leading to reduced proliferation rates, overexpression of exhaustion markers and increased susceptibility to Treg suppression and the influence of immune-modulating drugs such as rapamycin, FK506, and MMF. Notably, T cells cultured in low glucose concentrations increased the expression of BDH1 to utilize BHB as an alternative fuel source. Finally, the addition of BHB to the culture effectively rescued T cell impairments caused by insufficient glucose levels. CONCLUSIONS T cells display limited capacity to adapt to low glucose levels, resulting in profound functional impairment. However, T cell functions can be efficiently recovered by the presence of 2mM BHB.
Collapse
Affiliation(s)
- Arianna Ferrari
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Jessica Filoni
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Carla Di Dedda
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy
| | - Paolo Monti
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele Milan, Milan, Italy.
| |
Collapse
|
13
|
Bashir MA, Abdalla M, Shao CS, Wang H, Bondzie-Quaye P, Almahi WA, Swallah MS, Huang Q. Dual inhibitory potential of ganoderic acid A on GLUT1/3: computational and in vitro insights into targeting glucose metabolism in human lung cancer. RSC Adv 2024; 14:28569-28584. [PMID: 39247503 PMCID: PMC11378701 DOI: 10.1039/d4ra04454a] [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: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Human glucose transporters (GLUTs) facilitate the uptake of hexoses into cells. In cancer, the increased proliferation necessitates higher expression of GLUTs, with particular emphasis on GLUT1 and GLUT3. Thus, inhibiting GLUTs holds promise as an anticancer therapy by starving these cells of fuel. Ganoderic acid A (GAA), a triterpene found in Ganoderma lucidum, has anticancer and antidiabetic properties. Recent studies show that GAA reduces glucose uptake in cancer cells, which indicates that GAA may affect GLUT1/GLUT3 by inhibiting glucose uptake. Therefore, this study aimed to inspect whether GAA could target GLUT1/GLUT3 and play an inhibitory role in changing their endofacial and exofacial conformations. To this end, AlphaFold2 was employed to model the endofacial and exofacial conformations of GLUT3 and GLUT1, respectively. Molecular docking, molecular dynamics simulation, cell viability, cellular thermal shift assays (CETSA), glucose uptake, qPCR, and western blotting were harnessed. In comparison to the endofacial (cytochalasin B) and exofacial (phloretin) GLUT1/3 inhibitors, the computational findings unveiled GAA's capacity to bind and stabilize GLUT1/3 in their two conformational states, with a preference for binding the endofacial conformation. A low, non-cytotoxic dose of GAA thermally stabilized both transporters and inhibited glucose uptake in human lung cancer cells, similar to cytochalasin B and phloretin. In conclusion, this study has unearthed novel functionalities of GAA, suggesting its potential utility in cancer therapy by targeting glucose metabolism.
Collapse
Affiliation(s)
- Mona Alrasheed Bashir
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences Hefei 230031 China
- Science Island Branch of Graduate School, University of Science and Technology of China Hefei 230026 China
- Department of Biotechnology, Faculty of Science and Technology, Omdurman Islamic University P.O. Box 382 Omdurman Sudan
| | - Mohnad Abdalla
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University Jinan Shandong 250022 China
- Shandong Provincial Clinical Research Center for Children's Health and Disease Jinan Shandong 250022 China
| | - Chang-Sheng Shao
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences Hefei 230031 China
- Science Island Branch of Graduate School, University of Science and Technology of China Hefei 230026 China
| | - Han Wang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences Hefei 230031 China
- Science Island Branch of Graduate School, University of Science and Technology of China Hefei 230026 China
| | - Precious Bondzie-Quaye
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences Hefei 230031 China
- Science Island Branch of Graduate School, University of Science and Technology of China Hefei 230026 China
| | - Waleed Abdelbagi Almahi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences Hefei 230031 China
| | - Mohammed Sharif Swallah
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences Hefei 230031 China
- Science Island Branch of Graduate School, University of Science and Technology of China Hefei 230026 China
| | - Qing Huang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences Hefei 230031 China
- Science Island Branch of Graduate School, University of Science and Technology of China Hefei 230026 China
| |
Collapse
|
14
|
Colombo RB, Maxit C, Martinelli D, Anderson M, Masone D, Mayorga L. PURA and GLUT1: Sweet partners for brain health. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167261. [PMID: 38777099 DOI: 10.1016/j.bbadis.2024.167261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
PURA, also known as Pur-alpha, is an evolutionarily conserved DNA/RNA-binding protein crucial for various cellular processes, including DNA replication, transcriptional regulation, and translational control. Comprising three PUR domains, it engages with nucleic acids and has a role in protein-protein interactions. The manifestation of PURA syndrome, arising from mutations in the PURA gene, presents neurologically with developmental delay, hypotonia, and seizures. In our prior work from 2018, we highlighted the unique case of a PURA patient displaying hypoglycorrhachia, suggesting a potential association with GLUT1 dysfunction in this syndrome. In this current study, we expand the patient cohort with PURA mutations exhibiting hypoglycorrhachia and aim to unravel the molecular basis of this phenomenon. We established an in vitro model in HeLa cells to modulate PURA expression and investigated GLUT1 function and expression. Our findings indicate that PURA levels directly impact glucose uptake through the functioning of GLUT1, without influencing significantly GLUT1 expression. Moreover, our study reveals evidence for a possible physical interaction between PURA and GLUT1, demonstrated by colocalization and co-immunoprecipitation of both proteins. Computational analyses, employing molecular dynamics, further corroborates these findings, demonstrating that PURA:GLUT1 interactions are plausible, and that the stability of the complex is altered when PURA is truncated and/or mutated. In conclusion, our results suggest that PURA plays a pivotal role in driving the function of GLUT1 for glucose uptake, potentially forming a regulatory complex. Additional investigations are warranted to elucidate the precise mechanisms governing this complex and its significance in ensuring proper GLUT1 function.
Collapse
Affiliation(s)
- Rocío B Colombo
- Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina; Facultad De Química, Bioquímica y Farmacia, Universidad Nacional De San Luis, San Luis, Argentina
| | - Clarisa Maxit
- Servicio de Neurología infantil, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Diego Martinelli
- Division of Metabolism, Bambino Gesù Children's Hospital, Rome, Italy
| | - Mel Anderson
- PURA Foundation Australia, Plenty Victoria, Australia
| | - Diego Masone
- Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina; Facultad de Ingeniería, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| | - Lía Mayorga
- Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina; Instituto de Neurología Infantojuvenil (Neuroinfan), Mendoza, Argentina.
| |
Collapse
|
15
|
Toledano Zur R, Atar O, Barliya T, Hoogi S, Abramovich I, Gottlieb E, Ron-Harel N, Cohen CJ. Genetically engineering glycolysis in T cells increases their antitumor function. J Immunother Cancer 2024; 12:e008434. [PMID: 38964783 PMCID: PMC11227835 DOI: 10.1136/jitc-2023-008434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND T cells play a central role in the antitumor response. However, they often face numerous hurdles in the tumor microenvironment, including the scarcity of available essential metabolites such as glucose and amino acids. Moreover, cancer cells can monopolize these resources to thrive and proliferate by upregulating metabolite transporters and maintaining a high metabolic rate, thereby outcompeting T cells. METHODS Herein, we sought to improve T-cell antitumor function in the tumor vicinity by enhancing their glycolytic capacity to better compete with tumor cells. To achieve this, we engineered human T cells to express a key glycolysis enzyme, phosphofructokinase, in conjunction with Glucose transporter 3, a glucose transporter. We co-expressed these, along with tumor-specific chimeric antigen or T-cell receptors. RESULTS Engineered cells demonstrated an increased cytokine secretion and upregulation of T-cell activation markers compared with control cells. Moreover, they displayed superior glycolytic capacity, which translated into an improved in vivo therapeutic potential in a xenograft model of human tumors. CONCLUSION In summary, these findings support the implementation of T-cell metabolic engineering to enhance the efficacy of cellular immunotherapies for cancer.
Collapse
Affiliation(s)
| | - Orna Atar
- Technion Israel Institute of Technology, Haifa, Haifa, Israel
| | | | | | - Ifat Abramovich
- Technion Israel Institute of Technology, Haifa, Haifa, Israel
| | - Eyal Gottlieb
- Technion Israel Institute of Technology, Haifa, Haifa, Israel
| | - Noga Ron-Harel
- Technion Israel Institute of Technology, Haifa, Haifa, Israel
| | - Cyrille J Cohen
- Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Tel Aviv, Israel
| |
Collapse
|
16
|
Kim JH, Mailloux L, Bloor D, Tae H, Nguyen H, McDowell M, Padilla J, DeWaard A. Multiple roles for the cytoplasmic C-terminal domains of the yeast cell surface receptors Rgt2 and Snf3 in glucose sensing and signaling. Sci Rep 2024; 14:4055. [PMID: 38374219 PMCID: PMC10876965 DOI: 10.1038/s41598-024-54628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024] Open
Abstract
The plasma membrane proteins Rgt2 and Snf3 are glucose sensing receptors (GSRs) that generate an intracellular signal for the induction of gene expression in response to high and low extracellular glucose concentrations, respectively. The GSRs consist of a 12-transmembrane glucose recognition domain and a cytoplasmic C-terminal signaling tail. The GSR tails are dissimilar in length and sequence, but their distinct roles in glucose signal transduction are poorly understood. Here, we show that swapping the tails between Rgt2 and Snf3 does not alter the signaling activity of the GSRs, so long as their tails are phosphorylated in a Yck-dependent manner. Attachment of the GSR tails to Hxt1 converts the transporter into a glucose receptor; however, the tails attached to Hxt1 are not phosphorylated by the Ycks, resulting in only partial signaling. Moreover, in response to non-fermentable carbon substrates, Rgt2 and Hxt1-RT (RT, Rgt2-tail) are efficiently endocytosed, whereas Snf3 and Hxt1-ST (ST, Snf3-tail) are endocytosis-impaired. Thus, the tails are important regulatory domains required for the endocytosis of the Rgt2 and Snf3 glucose sensing receptors triggered by different cellular stimuli. Taken together, these results suggest multiple roles for the tail domains in GSR-mediated glucose sensing and signaling.
Collapse
Affiliation(s)
- Jeong-Ho Kim
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA.
| | - Levi Mailloux
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Daniel Bloor
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Haeun Tae
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Han Nguyen
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Morgan McDowell
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Jaqueline Padilla
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| | - Anna DeWaard
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA, 24502, USA
| |
Collapse
|
17
|
Kim JH, Mailloux L, Bloor D, Maddox B, Humble J. The role of salt bridge networks in the stability of the yeast hexose transporter 1. Biochim Biophys Acta Gen Subj 2023; 1867:130490. [PMID: 37844739 DOI: 10.1016/j.bbagen.2023.130490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND The yeast S. cerevisiae preferably metabolizes glucose through aerobic glycolysis. Glucose transport is facilitated by multiple hexose transporters (Hxts), and their expression and activity are tightly regulated by multiple mechanisms. However, detailed structural and functional analyses of Hxts remain limited, largely due to the lack of crystal structure. METHODS Homology modeling was used to build a 3D structural model for the yeast glucose transporter Hxt1 and investigate the effects of site directed mutations on Hxt1 stability and glucose transport activity. RESULTS The conserved salt bridge-forming residues observed in the human Glut4 and the yeast glucose receptor Rgt2 were identified within and between the two 6-transmembrane spanning segments of Hxt1. Most of the RGT2 mutations that disrupt the salt bridge networks were known to cause constitutive signal generation, whereas the corresponding substitutions in HXT1 were shown to decrease Hxt1 stability. While substitutions of the two residues in the salt bridge 2 in Glut4-E329Q and E393D-were reported to abolish glucose transport, the equivalent substitutions in Hxt1 (D382Q and E454D) did not affect Hxt1 glucose transport activity. CONCLUSIONS Substitutions of equivalent salt bridge-forming residues in Hxt1, Rgt2, and Glut4 are predicted to lock them in an inward-facing conformation but lead to different functional consequences. GENERAL SIGNIFICANCE The salt bridge networks in yeast and human glucose transporters and yeast glucose receptors may play different roles in maintaining their structural and functional integrity.
Collapse
Affiliation(s)
- Jeong-Ho Kim
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA.
| | - Levi Mailloux
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA
| | - Daniel Bloor
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA
| | - Bradley Maddox
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA
| | - Julia Humble
- Department of Biology and Chemistry, Liberty University, 1971 University Blvd, Lynchburg, VA 24502, USA
| |
Collapse
|
18
|
Farjadian F, Faghih Z, Fakhimi M, Iranpour P, Mohammadi-Samani S, Doroudian M. Glucosamine-Modified Mesoporous Silica-Coated Magnetic Nanoparticles: A "Raisin-Cake"-like Structure as an Efficient Theranostic Platform for Targeted Methotrexate Delivery. Pharmaceutics 2023; 15:2491. [PMID: 37896251 PMCID: PMC10610088 DOI: 10.3390/pharmaceutics15102491] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
This study presents the synthesis of glucosamine-modified mesoporous silica-coated magnetic nanoparticles (MNPs) as a therapeutic platform for the delivery of an anticancer drug, methotrexate (MTX). The MNPs were coated with mesoporous silica in a templated sol-gel process to form MNP@MSN, and then chloropropyl groups were added to the structure in a post-modification reaction. Glucosamine was then reacted with the chloro-modified structure, and methotrexate was conjugated to the hydroxyl group of the glucose. The prepared structure was characterized using techniques such as Fourier transform infrared (FT-IR) spectroscopy, elemental analysis (CHN), field emission scanning electron microscopy (FESEM), transmission electron microscopy (TEM), dynamic light scattering (DLS), a vibrating sample magnetometer (VSM), and X-ray diffraction (XRD). Good formation of nano-sized MNPs and MNP@MSN was observed via particle size monitoring. The modified glucosamine structure showed a controlled release profile of methotrexate in simulated tumor fluid. In vitro evaluation using the 4T1 breast cancer cell line showed the cytotoxicity, apoptosis, and cell cycle effects of methotrexate. The MTT assay showed comparable toxicity between MTX-loaded nanoparticles and free MTX. The structure could act as a glucose transporter-targeting agent and showed increased uptake in cancer cells. An in vivo breast cancer model was established in BALB/C mice, and the distribution of MTX-conjugated MNP@MSN particles was visualized using MRI. The MTX-conjugated particles showed significant anti-tumor potential together with MRI contrast enhancement.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Canter, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran;
| | - Zahra Faghih
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45550, Iran; (Z.F.); (M.F.)
| | - Maryam Fakhimi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45550, Iran; (Z.F.); (M.F.)
| | - Pooya Iranpour
- Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz 71936-13311, Iran;
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Canter, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran;
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71468-64685, Iran
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 15719-14911, Iran
| |
Collapse
|
19
|
Zeng YC, Sobti M, Quinn A, Smith NJ, Brown SHJ, Vandenberg JI, Ryan RM, O'Mara ML, Stewart AG. Structural basis of promiscuous substrate transport by Organic Cation Transporter 1. Nat Commun 2023; 14:6374. [PMID: 37821493 PMCID: PMC10567722 DOI: 10.1038/s41467-023-42086-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023] Open
Abstract
Organic Cation Transporter 1 (OCT1) plays a crucial role in hepatic metabolism by mediating the uptake of a range of metabolites and drugs. Genetic variations can alter the efficacy and safety of compounds transported by OCT1, such as those used for cardiovascular, oncological, and psychological indications. Despite its importance in drug pharmacokinetics, the substrate selectivity and underlying structural mechanisms of OCT1 remain poorly understood. Here, we present cryo-EM structures of full-length human OCT1 in the inward-open conformation, both ligand-free and drug-bound, indicating the basis for its broad substrate recognition. Comparison of our structures with those of outward-open OCTs provides molecular insight into the alternating access mechanism of OCTs. We observe that hydrophobic gates stabilize the inward-facing conformation, whereas charge neutralization in the binding pocket facilitates the release of cationic substrates. These findings provide a framework for understanding the structural basis of the promiscuity of drug binding and substrate translocation in OCT1.
Collapse
Affiliation(s)
- Yi C Zeng
- Molecular, Structural and Computational Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| | - Meghna Sobti
- Molecular, Structural and Computational Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Ada Quinn
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Nicola J Smith
- School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW, Australia
| | - Simon H J Brown
- School of Chemistry and Molecular Bioscience, Molecular Horizons, and Australian Research Council Centre for Cryo-electron Microscopy of Membrane Proteins, University of Wollongong, Wollongong, NSW, Australia
| | - Jamie I Vandenberg
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
- Molecular Cardiology and Biophysics Division, The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Renae M Ryan
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Megan L O'Mara
- Australian Institute of Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Alastair G Stewart
- Molecular, Structural and Computational Biology Division, The Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
20
|
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
|
21
|
Olszańska J, Pietraszek-Gremplewicz K, Domagalski M, Nowak D. Mutual impact of adipocytes and colorectal cancer cells growing in co-culture conditions. Cell Commun Signal 2023; 21:130. [PMID: 37316878 DOI: 10.1186/s12964-023-01155-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/29/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common malignancy worldwide. CRC cells are situated in an adipocyte-rich microenvironment, which leads to interactions between adipocytes and CRC cells. Upon exposure to cancer cells, adipocytes transform into cancer-associated adipocytes (CAAs), and as a result, they gain features that promote tumor progression. The aim of this research was to shed more light on the detailed role of interactions between adipocytes and CRC cells associated with cancer progression in the context of these alterations. METHODS To implement adipocyte-CRC cell interaction, a co-culture model was applied. The analyses mainly focused on the metabolic modifications within CAAs and CRC cells, as well as the proliferation and migration potential of CRC cells. The impact of CRC on adipocytes was investigated by qRT-PCR analysis and Oil Red O staining. Proliferation and migration of CRC cells upon co-culture were tested with videomicroscopy, XTT, and a wound healing assay. Metabolic changes within CAAs and CRC cells were investigated based on lipid droplet formation, cell cycle analysis, gene and protein expression by qRT-PCR, and western blotting techniques. RESULTS CRC cells induced reprogramming of adipocytes into CAAs, which was connected with downregulation of lipid droplet formation in CAAs and alteration in adipocyte features. CAAs showed decreased metabolism-related gene expression, phosphorylation of Akt, ERK kinases, STAT3, and lactate secretion in comparison to the control. CAAs also promoted the migration, proliferation, and lipid droplet accumulation of CRC cells. After co-culturing with adipocytes, there was a shift to the G2/M phase of the cell cycle according to the differences in cyclin expression. CONCLUSION There are complex bidirectional interactions between adipocytes and CRC cells that may be connected with the induction of CRC cell progression. Video Abstract.
Collapse
Affiliation(s)
- Joanna Olszańska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | | | - Mikołaj Domagalski
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| |
Collapse
|
22
|
Bavnhøj L, Driller JH, Zuzic L, Stange AD, Schiøtt B, Pedersen BP. Structure and sucrose binding mechanism of the plant SUC1 sucrose transporter. NATURE PLANTS 2023; 9:938-950. [PMID: 37188854 PMCID: PMC10281868 DOI: 10.1038/s41477-023-01421-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/19/2023] [Indexed: 05/17/2023]
Abstract
Sucrose import from photosynthetic tissues into the phloem is mediated by transporters from the low-affinity sucrose transporter family (SUC/SUT family). Furthermore, sucrose redistribution to other tissues is driven by phloem sap movement, the product of high turgor pressure created by this import activity. Additionally, sink organs such as fruits, cereals and seeds that accumulate high concentrations of sugar also depend on this active transport of sucrose. Here we present the structure of the sucrose-proton symporter, Arabidopsis thaliana SUC1, in an outward open conformation at 2.7 Å resolution, together with molecular dynamics simulations and biochemical characterization. We identify the key acidic residue required for proton-driven sucrose uptake and describe how protonation and sucrose binding are strongly coupled. Sucrose binding is a two-step process, with initial recognition mediated by the glucosyl moiety binding directly to the key acidic residue in a stringent pH-dependent manner. Our results explain how low-affinity sucrose transport is achieved in plants, and pinpoint a range of SUC binders that help define selectivity. Our data demonstrate a new mode for proton-driven symport with links to cation-driven symport and provide a broad model for general low-affinity transport in highly enriched substrate environments.
Collapse
Affiliation(s)
- Laust Bavnhøj
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan Heiner Driller
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Lorena Zuzic
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | | | - Birgit Schiøtt
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
23
|
Milne RJ, Dibley KE, Bose J, Ashton AR, Ryan PR, Tyerman SD, Lagudah ES. Expression of the wheat multipathogen resistance hexose transporter Lr67res is associated with anion fluxes. PLANT PHYSIOLOGY 2023; 192:1254-1267. [PMID: 36806945 DOI: 10.1093/plphys/kiad104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/24/2023] [Accepted: 01/29/2023] [Indexed: 06/01/2023]
Abstract
Many disease resistance genes in wheat (Triticum aestivum L.) confer strong resistance to specific pathogen races or strains, and only a small number of genes confer multipathogen resistance. The Leaf rust resistance 67 (Lr67) gene fits into the latter category as it confers partial resistance to multiple biotrophic fungal pathogens in wheat and encodes a Sugar Transport Protein 13 (STP13) family hexose-proton symporter variant. Two mutations (G144R, V387L) in the resistant variant, Lr67res, differentiate it from the susceptible Lr67sus variant. The molecular function of the Lr67res protein is not understood, and this study aimed to broaden our knowledge on this topic. Biophysical analysis of the wheat Lr67sus and Lr67res protein variants was performed using Xenopus laevis oocytes as a heterologous expression system. Oocytes injected with Lr67sus displayed properties typically associated with proton-coupled sugar transport proteins-glucose-dependent inward currents, a Km of 110 ± 10 µM glucose, and a substrate selectivity permitting the transport of pentoses and hexoses. By contrast, Lr67res induced much larger sugar-independent inward currents in oocytes, implicating an alternative function. Since Lr67res is a mutated hexose-proton symporter, the possibility of protons underlying these currents was investigated but rejected. Instead, currents in Lr67res oocytes appeared to be dominated by anions. This conclusion was supported by electrophysiology and 36Cl- uptake studies and the similarities with oocytes expressing the known chloride channel from Torpedo marmorata, TmClC-0. This study provides insights into the function of an important disease resistance gene in wheat, which can be used to determine how this gene variant underpins disease resistance in planta.
Collapse
Affiliation(s)
- Ricky J Milne
- CSIRO, Agriculture and Food, Canberra, ACT 2601, Australia
| | | | - Jayakumar Bose
- Australian Research Council Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine, University of Adelaide, Urrbrae, SA 5064, Australia
- School of Science, Western Sydney University, Richmond, NSW 2753, Australia
| | | | - Peter R Ryan
- CSIRO, Agriculture and Food, Canberra, ACT 2601, Australia
| | - Stephen D Tyerman
- Australian Research Council Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine, University of Adelaide, Urrbrae, SA 5064, Australia
| | | |
Collapse
|
24
|
Todorova V, Stauffacher MF, Ravotto L, Nötzli S, Karademir D, Ebner LJA, Imsand C, Merolla L, Hauck SM, Samardzija M, Saab AS, Barros LF, Weber B, Grimm C. Deficits in mitochondrial TCA cycle and OXPHOS precede rod photoreceptor degeneration during chronic HIF activation. Mol Neurodegener 2023; 18:15. [PMID: 36882871 PMCID: PMC9990367 DOI: 10.1186/s13024-023-00602-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/03/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Major retinal degenerative diseases, including age-related macular degeneration, diabetic retinopathy and retinal detachment, are associated with a local decrease in oxygen availability causing the formation of hypoxic areas affecting the photoreceptor (PR) cells. Here, we addressed the underlying pathological mechanisms of PR degeneration by focusing on energy metabolism during chronic activation of hypoxia-inducible factors (HIFs) in rod PR. METHODS We used two-photon laser scanning microscopy (TPLSM) of genetically encoded biosensors delivered by adeno-associated viruses (AAV) to determine lactate and glucose dynamics in PR and inner retinal cells. Retinal layer-specific proteomics, in situ enzymatic assays and immunofluorescence studies were used to analyse mitochondrial metabolism in rod PRs during chronic HIF activation. RESULTS PRs exhibited remarkably higher glycolytic flux through the hexokinases than neurons of the inner retina. Chronic HIF activation in rods did not cause overt change in glucose dynamics but an increase in lactate production nonetheless. Furthermore, dysregulation of the oxidative phosphorylation pathway (OXPHOS) and tricarboxylic acid (TCA) cycle in rods with an activated hypoxic response decelerated cellular anabolism causing shortening of rod photoreceptor outer segments (OS) before onset of cell degeneration. Interestingly, rods with deficient OXPHOS but an intact TCA cycle did not exhibit these early signs of anabolic dysregulation and showed a slower course of degeneration. CONCLUSION Together, these data indicate an exceeding high glycolytic flux in rods and highlight the importance of mitochondrial metabolism and especially of the TCA cycle for PR survival in conditions of increased HIF activity.
Collapse
Affiliation(s)
- Vyara Todorova
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Mia Fee Stauffacher
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology and Neuroscience Center Zurich, University and ETH Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland
| | - Sarah Nötzli
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Duygu Karademir
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Lynn J A Ebner
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Cornelia Imsand
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Luca Merolla
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Marijana Samardzija
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology and Neuroscience Center Zurich, University and ETH Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland
| | - L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile.,Universidad San Sebastián, Valdivia, Chile
| | - Bruno Weber
- Institute of Pharmacology and Toxicology and Neuroscience Center Zurich, University and ETH Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland
| | - Christian Grimm
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland.
| |
Collapse
|
25
|
Wu WZ, Bai YP. Endothelial GLUTs and vascular biology. Biomed Pharmacother 2023; 158:114151. [PMID: 36565587 DOI: 10.1016/j.biopha.2022.114151] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Endothelial metabolism is a promising target for vascular functional regulation and disease therapy. Glucose is the primary fuel for endothelial metabolism, supporting ATP generation and endothelial cell survival. Multiple studies have discussed the role of endothelial glucose catabolism, such as glycolysis and oxidative phosphorylation, in vascular functional remodeling. However, the role of the first gatekeepers of endothelial glucose utilization, glucose transporters, in the vasculature has long been neglected. Here, this review summarizes glucose transporter studies in vascular research. We mainly focus on GLUT1 and GLUT3 because they are the most critical glucose transporters responsible for most endothelial glucose uptake. Some interesting topics are also discussed, intending to provide directions for endothelial glucose transporter research in the future.
Collapse
Affiliation(s)
- Wan-Zhou Wu
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Center for Vascular Disease and Translational Medicine, Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Ping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
26
|
Platinum glycoconjugates: "Sweet bullets" for targeted cancer therapy? Curr Opin Chem Biol 2023; 72:102236. [PMID: 36516491 DOI: 10.1016/j.cbpa.2022.102236] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022]
Abstract
Cancer, which is characterized by uncontrolled proliferation of abnormal cells, is a leading cause of morbidity and mortality worldwide. Cytotoxic chemotherapy, especially with platinum drugs, remains the mainstay of cancer treatment in the clinical setting. Despite phenomenal success, small-molecule chemotherapeutic drugs suffer from some serious drawbacks. Lack of cancer selectivity and the ensuing side effects mar the therapeutic potential of these drugs. Glycoconjugation has emerged as an attractive strategy for imparting selectivity and improving pharmacokinetics of cytotoxic agents. In this review, we provide an overview of the glycoconjugation strategy and then illustrate the application of this strategy with the help of some concrete examples of platinum based glycoconjugates. At the end we discuss a few important aspects of these glycoconjugates which merit further investigations.
Collapse
|
27
|
Identification of Structural Determinants of the Transport of the Dehydroascorbic Acid Mediated by Glucose Transport GLUT1. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020521. [PMID: 36677580 PMCID: PMC9867014 DOI: 10.3390/molecules28020521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/12/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023]
Abstract
GLUT1 is a facilitative glucose transporter that can transport oxidized vitamin C (i.e., dehydroascorbic acid) and complements the action of reduced vitamin C transporters. To identify the residues involved in human GLUT1's transport of dehydroascorbic acid, we performed docking studies in the 5 Å grid of the glucose-binding cavity of GLUT1. The interactions of the bicyclic hemiacetal form of dehydroascorbic acid with GLUT1 through hydrogen bonds with the -OH group of C3 and C5 were less favorable than the interactions with the sugars transported by GLUT1. The eight most relevant residues in such interactions (i.e., F26, Q161, I164, Q282, Y292, and W412) were mutated to alanine to perform functional studies for dehydroascorbic acid and the glucose analog, 2-deoxiglucose, in Xenopus laevis oocytes. All the mutants decreased the uptake of both substrates to less than 50%. The partial effect of the N317A mutant in transporting dehydroascorbic acid was associated with a 30% decrease in the Vmax compared to the wildtype GLUT1. The results show that both substrates share the eight residues studied in GLUT1, albeit with a differential contribution of N317. Our work, combining docking with functional studies, marks the first to identify structural determinants of oxidized vitamin C's transport via GLUT1.
Collapse
|
28
|
Tiemann JKS, Zschach H, Lindorff-Larsen K, Stein A. Interpreting the molecular mechanisms of disease variants in human transmembrane proteins. Biophys J 2023:S0006-3495(22)03941-8. [PMID: 36600598 DOI: 10.1016/j.bpj.2022.12.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/19/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
Next-generation sequencing of human genomes reveals millions of missense variants, some of which may lead to loss of protein function and ultimately disease. Here, we investigate missense variants in membrane proteins-key drivers in cell signaling and recognition. We find enrichment of pathogenic variants in the transmembrane region across 19,000 functionally classified variants in human membrane proteins. To accurately predict variant consequences, one fundamentally needs to understand the underlying molecular processes. A key mechanism underlying pathogenicity in missense variants of soluble proteins has been shown to be loss of stability. Membrane proteins, however, are widely understudied. Here, we interpret variant effects on a larger scale by performing structure-based estimations of changes in thermodynamic stability using a membrane-specific energy function and analyses of sequence conservation during evolution of 15 transmembrane proteins. We find evidence for loss of stability being the cause of pathogenicity in more than half of the pathogenic variants, indicating that this is a driving factor also in membrane-protein-associated diseases. Our findings show how computational tools aid in gaining mechanistic insights into variant consequences for membrane proteins. To enable broader analyses of disease-related and population variants, we include variant mappings for the entire human proteome.
Collapse
Affiliation(s)
- Johanna Katarina Sofie Tiemann
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Henrike Zschach
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Amelie Stein
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
29
|
Mauri A, Duse A, Palm G, Previtali R, Bova SM, Olivotto S, Benedetti S, Coscia F, Veggiotti P, Cereda C. Molecular Genetics of GLUT1DS Italian Pediatric Cohort: 10 Novel Disease-Related Variants and Structural Analysis. Int J Mol Sci 2022; 23:ijms232113560. [PMID: 36362347 PMCID: PMC9654628 DOI: 10.3390/ijms232113560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022] Open
Abstract
GLUT1 deficiency syndrome (GLUT1DS1; OMIM #606777) is a rare genetic metabolic disease, characterized by infantile-onset epileptic encephalopathy, global developmental delay, progressive microcephaly, and movement disorders (e.g., spasticity and dystonia). It is caused by heterozygous mutations in the SLC2A1 gene, which encodes the GLUT1 protein, a glucose transporter across the blood-brain barrier (BBB). Most commonly, these variants arise de novo resulting in sporadic cases, although several familial cases with AD inheritance pattern have been described. Twenty-seven Italian pediatric patients, clinically suspect of GLUT1DS from both sporadic and familial cases, have been enrolled. We detected by trios sequencing analysis 25 different variants causing GLUT1DS. Of these, 40% of the identified variants (10 out of 25) had never been reported before, including missense, frameshift, and splice site variants. Their structural mapping on the X-ray structure of GLUT1 strongly suggested the potential pathogenic effects of these novel disease-related mutations, broadening the genotypic spectrum heterogeneity found in the SLC2A1 gene. Moreover, 24% is located in a vulnerable region of the GLUT1 protein that involves transmembrane 4 and 5 helices encoded by exon 4, confirming a mutational hotspot in the SLC2A1 gene. Lastly, we investigated possible correlations between mutation type and clinical and biochemical data observed in our GLUT1DS cohort, revealing that splice site and frameshift variants are related to a more severe phenotype and low CSF parameters.
Collapse
Affiliation(s)
- Alessia Mauri
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy
- Newborn Screening and Genetic Metabolic Diseases Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Alessandra Duse
- Pediatric Neurology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Giacomo Palm
- Structural Biology Center, Human Technopole, 20157 Milan, Italy
| | - Roberto Previtali
- Pediatric Neurology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | | | - Sara Olivotto
- Pediatric Neurology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Sara Benedetti
- Newborn Screening and Genetic Metabolic Diseases Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | | | - Pierangelo Veggiotti
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy
- Pediatric Neurology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Cristina Cereda
- Newborn Screening and Genetic Metabolic Diseases Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
- Correspondence:
| |
Collapse
|
30
|
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
|
31
|
von Eugen K, Endepols H, Drzezga A, Neumaier B, Güntürkün O, Backes H, Ströckens F. Avian neurons consume three times less glucose than mammalian neurons. Curr Biol 2022; 32:4306-4313.e4. [PMID: 36084646 DOI: 10.1016/j.cub.2022.07.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/11/2022] [Accepted: 07/26/2022] [Indexed: 12/14/2022]
Abstract
Brains are among the most energetically costly tissues in the mammalian body.1 This is predominantly caused by expensive neurons with high glucose demands.2 Across mammals, the neuronal energy budget appears to be fixed, possibly posing an evolutionary constraint on brain growth.3-6 Compared to similarly sized mammals, birds have higher numbers of neurons, and this advantage conceivably contributes to their cognitive prowess.7 We set out to determine the neuronal energy budget of birds to elucidate how they can metabolically support such high numbers of neurons. We estimated glucose metabolism using positron emission tomography (PET) and 2-[18F]fluoro-2-deoxyglucose ([18F]FDG) as the radiotracer in awake and anesthetized pigeons. Combined with kinetic modeling, this is the gold standard to quantify cerebral metabolic rate of glucose consumption (CMRglc).8 We found that neural tissue in the pigeon consumes 27.29 ± 1.57 μmol glucose per 100 g per min in an awake state, which translates into a surprisingly low neuronal energy budget of 1.86 × 10-9 ± 0.2 × 10-9 μmol glucose per neuron per minute. This is approximately 3 times lower than the rate in the average mammalian neuron.3 The remarkably low neuronal energy budget explains how pigeons, and possibly other avian species, can support such high numbers of neurons without associated metabolic costs or compromising neuronal signaling. The advantage in neuronal processing of information at a higher efficiency possibly emerged during the distinct evolution of the avian brain.
Collapse
Affiliation(s)
- Kaya von Eugen
- Department of Biopsychology, Institute of Cognitive Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Heike Endepols
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany; Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine, INM-5: Nuclear Chemistry, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine, INM-2: Molecular Organization of the Brain, Forschungszentrum Jülich GmbH, Jülich, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine, INM-5: Nuclear Chemistry, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Onur Güntürkün
- Department of Biopsychology, Institute of Cognitive Neuroscience, Ruhr University Bochum, Bochum, Germany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Multimodal Imaging Group, Cologne, Germany
| | - Felix Ströckens
- Department of Biopsychology, Institute of Cognitive Neuroscience, Ruhr University Bochum, Bochum, Germany; Cécile and Oskar Vogt Institute of Brain Research, University Hospital Düsseldorf, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
32
|
Owoloye AJ, Ligali FC, Enejoh OA, Musa AZ, Aina O, Idowu ET, Oyebola KM. Molecular docking, simulation and binding free energy analysis of small molecules as PfHT1 inhibitors. PLoS One 2022; 17:e0268269. [PMID: 36026508 PMCID: PMC9417013 DOI: 10.1371/journal.pone.0268269] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/02/2022] [Indexed: 11/19/2022] Open
Abstract
Antimalarial drug resistance has thrown a spanner in the works of malaria elimination. New drugs are required for ancillary support of existing malaria control efforts. Plasmodium falciparum requires host glucose for survival and proliferation. On this basis, P. falciparum hexose transporter 1 (PfHT1) protein involved in hexose permeation is considered a potential drug target. In this study, we tested the antimalarial activity of some compounds against PfHT1 using computational techniques. We performed high throughput virtual screening of 21,352 small-molecule compounds against PfHT1. The stability of the lead compound complexes was evaluated via molecular dynamics (MD) simulation for 100 nanoseconds. We also investigated the pharmacodynamic, pharmacokinetic and physiological characteristics of the compounds in accordance with Lipinksi rules for drug-likeness to bind and inhibit PfHT1. Molecular docking and free binding energy analyses were carried out using Molecular Mechanics with Generalized Born and Surface Area (MMGBSA) solvation to determine the selectivity of the hit compounds for PfHT1 over the human glucose transporter (hGLUT1) orthologue. Five important PfHT1 inhibitors were identified: Hyperoside (CID5281643); avicularin (CID5490064); sylibin (CID5213); harpagoside (CID5481542) and quercetagetin (CID5281680). The compounds formed intermolecular interaction with the binding pocket of the PfHT1 target via conserved amino acid residues (Val314, Gly183, Thr49, Asn52, Gly183, Ser315, Ser317, and Asn48). The MMGBSA analysis of the complexes yielded high free binding energies. Four (CID5281643, CID5490064, CID5213, and CID5481542) of the identified compounds were found to be stable within the PfHT1 binding pocket throughout the 100 nanoseconds simulation run time. The four compounds demonstrated higher affinity for PfHT1 than the human major glucose transporter (hGLUT1). This investigation demonstrates the inhibition potential of sylibin, hyperoside, harpagoside, and avicularin against PfHT1 receptor. Robust preclinical investigations are required to validate the chemotherapeutic properties of the identified compounds.
Collapse
Affiliation(s)
- Afolabi J. Owoloye
- Center for Genomic Research in Biomedicine (CeGRIB), College of Basic and Applied Sciences, Mountain Top University, Ibafo, Nigeria
- Parasitology and Bioinformatics Unit, Department of Zoology, Faculty of Science, University of Lagos, Lagos, Nigeria
- Nigerian Institute of Medical Research, Lagos, Nigeria
| | - Funmilayo C. Ligali
- Nigerian Institute of Medical Research, Lagos, Nigeria
- Biochemistry Department, Faculty of Basic Medical Science, University of Lagos, Lagos, Nigeria
| | - Ojochenemi A. Enejoh
- Genetics, Genomics and Bioinformatics Department, National Biotechnology Development Agency, Abuja, Nigeria
| | | | | | - Emmanuel T. Idowu
- Parasitology and Bioinformatics Unit, Department of Zoology, Faculty of Science, University of Lagos, Lagos, Nigeria
| | - Kolapo M. Oyebola
- Center for Genomic Research in Biomedicine (CeGRIB), College of Basic and Applied Sciences, Mountain Top University, Ibafo, Nigeria
- Nigerian Institute of Medical Research, Lagos, Nigeria
| |
Collapse
|
33
|
Abstract
Linus Pauling in 1950 published a three-dimensional model for a universal protein secondary structure motif which he initially called the alpha-spiral. Jack Dunitz, then a postdoc in Pauling's lab suggested to Pauling that the term helix is more accurate than spiral when describing the right-handed peptide and protein coiled structures. Pauling agreed, hence the rise of the alpha-helix, and, by extension, the ‘double helix’ structure of DNA. Although structural biologists and protein chemists are familiar with varying polar and apolar characters of amino acids in alpha-helices, to non-experts the three chemically distinct alpha-helix types classified here may hide in plain sight.
Collapse
Affiliation(s)
- Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Martin Egli
- Department of Biochemistry, Vanderbilt University, School of Medicine, Nashville, Tennessee 37232-0146, USA
| |
Collapse
|
34
|
One Molecule for Mental Nourishment and More: Glucose Transporter Type 1—Biology and Deficiency Syndrome. Biomedicines 2022; 10:biomedicines10061249. [PMID: 35740271 PMCID: PMC9219734 DOI: 10.3390/biomedicines10061249] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
Glucose transporter type 1 (Glut1) is the main transporter involved in the cellular uptake of glucose into many tissues, and is highly expressed in the brain and in erythrocytes. Glut1 deficiency syndrome is caused mainly by mutations of the SLC2A1 gene, impairing passive glucose transport across the blood–brain barrier. All age groups, from infants to adults, may be affected, with age-specific symptoms. In its classic form, the syndrome presents as an early-onset drug-resistant metabolic epileptic encephalopathy with a complex movement disorder and developmental delay. In later-onset forms, complex motor disorder predominates, with dystonia, ataxia, chorea or spasticity, often triggered by fasting. Diagnosis is confirmed by hypoglycorrhachia (below 45 mg/dL) with normal blood glucose, 18F-fluorodeoxyglucose positron emission tomography, and genetic analysis showing pathogenic SLC2A1 variants. There are also ongoing positive studies on erythrocytes’ Glut1 surface expression using flow cytometry. The standard treatment still consists of ketogenic therapies supplying ketones as alternative brain fuel. Anaplerotic substances may provide alternative energy sources. Understanding the complex interactions of Glut1 with other tissues, its signaling function for brain angiogenesis and gliosis, and the complex regulation of glucose transportation, including compensatory mechanisms in different tissues, will hopefully advance therapy. Ongoing research for future interventions is focusing on small molecules to restore Glut1, metabolic stimulation, and SLC2A1 transfer strategies. Newborn screening, early identification and treatment could minimize the neurodevelopmental disease consequences. Furthermore, understanding Glut1 relative deficiency or inhibition in inflammation, neurodegenerative disorders, and viral infections including COVID-19 and other settings could provide clues for future therapeutic approaches.
Collapse
|
35
|
Insights into the structure and function of the human organic anion transporter 1 in lipid bilayer membranes. Sci Rep 2022; 12:7057. [PMID: 35488116 PMCID: PMC9054760 DOI: 10.1038/s41598-022-10755-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/12/2022] [Indexed: 01/27/2023] Open
Abstract
The human SLC22A6/OAT1 plays an important role in the elimination of a broad range of endogenous substances and xenobiotics thus attracting attention from the pharmacological community. Furthermore, OAT1 is also involved in key physiological events such as the remote inter-organ communication. Despite its significance, the knowledge about hOAT1 structure and the transport mechanism at the atomic level remains fragmented owing to the lack of resolved structures. By means of protein-threading modeling refined by μs-scaled Molecular Dynamics simulations, the present study provides the first robust model of hOAT1 in outward-facing conformation. Taking advantage of the AlphaFold 2 predicted structure of hOAT1 in inward-facing conformation, we here provide the essential structural and functional features comparing both states. The intracellular motifs conserved among Major Facilitator Superfamily members create a so-called “charge-relay system” that works as molecular switches modulating the conformation. The principal element of the event points at interactions of charged residues that appear crucial for the transporter dynamics and function. Moreover, hOAT1 model was embedded in different lipid bilayer membranes highlighting the crucial structural dependence on lipid-protein interactions. MD simulations supported the pivotal role of phosphatidylethanolamine components to the protein conformation stability. The present model is made available to decipher the impact of any observed polymorphism and mutation on drug transport as well as to understand substrate binding modes.
Collapse
|
36
|
Ryniawec JM, Coope MR, Loertscher E, Bageerathan V, de Oliveira Pessoa D, Warfel NA, Cress AE, Padi M, Rogers GC. GLUT3/SLC2A3 Is an Endogenous Marker of Hypoxia in Prostate Cancer Cell Lines and Patient-Derived Xenograft Tumors. Diagnostics (Basel) 2022; 12:diagnostics12030676. [PMID: 35328229 PMCID: PMC8946944 DOI: 10.3390/diagnostics12030676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 01/17/2023] Open
Abstract
The microenvironment of solid tumors is dynamic and frequently contains pockets of low oxygen levels (hypoxia) surrounded by oxygenated tissue. Indeed, a compromised vasculature is a hallmark of the tumor microenvironment, creating both spatial gradients and temporal variability in oxygen availability. Notably, hypoxia associates with increased metastasis and poor survival in patients. Therefore, to aid therapeutic decisions and better understand hypoxia’s role in cancer progression, it is critical to identify endogenous biomarkers of hypoxia to spatially phenotype oncogenic lesions in human tissue, whether precancerous, benign, or malignant. Here, we characterize the glucose transporter GLUT3/SLC2A3 as a biomarker of hypoxic prostate epithelial cells and prostate tumors. Transcriptomic analyses of non-tumorigenic, immortalized prostate epithelial cells revealed a highly significant increase in GLUT3 expression under hypoxia. Additionally, GLUT3 protein increased 2.4-fold in cultured hypoxic prostate cell lines and was upregulated within hypoxic regions of xenograft tumors, including two patient-derived xenografts (PDX). Finally, GLUT3 out-performs other established hypoxia markers; GLUT3 staining in PDX specimens detects 2.6–8.3 times more tumor area compared to a mixture of GLUT1 and CA9 antibodies. Therefore, given the heterogeneous nature of tumors, we propose adding GLUT3 to immunostaining panels when trying to detect hypoxic regions in prostate samples.
Collapse
Affiliation(s)
- John M. Ryniawec
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719, USA; (J.M.R.); (M.R.C.); (E.L.); (N.A.W.)
| | - Matthew R. Coope
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719, USA; (J.M.R.); (M.R.C.); (E.L.); (N.A.W.)
| | - Emily Loertscher
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719, USA; (J.M.R.); (M.R.C.); (E.L.); (N.A.W.)
| | - Vignesh Bageerathan
- Biostatistics and Bioinformatics Shared Resource, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (V.B.); (D.d.O.P.)
| | - Diogo de Oliveira Pessoa
- Biostatistics and Bioinformatics Shared Resource, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA; (V.B.); (D.d.O.P.)
| | - Noel A. Warfel
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719, USA; (J.M.R.); (M.R.C.); (E.L.); (N.A.W.)
| | - Anne E. Cress
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719, USA; (J.M.R.); (M.R.C.); (E.L.); (N.A.W.)
- Correspondence: (A.E.C.); (M.P.); (G.C.R.)
| | - Megha Padi
- Department of Molecular and Cellular Biology, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
- Correspondence: (A.E.C.); (M.P.); (G.C.R.)
| | - Gregory C. Rogers
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719, USA; (J.M.R.); (M.R.C.); (E.L.); (N.A.W.)
- Correspondence: (A.E.C.); (M.P.); (G.C.R.)
| |
Collapse
|
37
|
Comparing 2 crystal structures and 12 AlphaFold2-predicted human membrane glucose transporters and their water-soluble glutamine, threonine and tyrosine variants. QRB DISCOVERY 2022. [PMID: 37529287 PMCID: PMC10392618 DOI: 10.1017/qrd.2022.6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Membrane transporters including glucose transporters (GLUTs) are involved in cellular energy supplies, cell metabolism and other vital biological activities. They have also been implicated in cancer proliferation and metastasis, thus they represent an important target in combatting cancer. However, membrane transporters are very difficult to study due to their multispan transmembrane properties. The new computational tool, AlphaFold2, offers highly accurate predictions of three-dimensional protein structures. The glutamine, threonine and tyrosine (QTY) code provides a systematic method of rendering hydrophobic sequences into hydrophilic ones. Here, we present computational studies of native integral membrane GLUTs with 12 transmembrane helical segments determined by X-ray crystallography and CryoEM, comparing the AlphaFold2-predicted native structure to their water-soluble QTY variants predicted by AlphaFold2. In the native structures of the transmembrane helices, there are hydrophobic amino acids leucine (L), isoleucine (I), valine (V) and phenylalanine (F). Applying the QTY code, these hydrophobic amino acids are systematically replaced by hydrophilic amino acids, glutamine (Q), threonine (T) and tyrosine (Y) rendering them water-soluble. We present the superposed structures of native GLUTs and their water-soluble QTY variants. The superposed structures show remarkable similar residue mean square distance values between 0.47 and 3.6 Å (most about 1–2 Å) despite >44% transmembrane amino acid differences. We also show the differences of hydrophobicity patches between the native membrane transporters and their QTY variants. We explain the rationale why the membrane protein QTY variants become water-soluble. Our study provides insight into the differences between the hydrophobic helices and hydrophilic helices, and offers confirmation of the QTY method for studying multispan transmembrane proteins and other aggregated proteins through their water-soluble variants.
Collapse
|
38
|
Udrea AM, Gradisteanu Pircalabioru G, Boboc AA, Mares C, Dinache A, Mernea M, Avram S. Advanced Bioinformatics Tools in the Pharmacokinetic Profiles of Natural and Synthetic Compounds with Anti-Diabetic Activity. Biomolecules 2021; 11:1692. [PMID: 34827690 PMCID: PMC8615418 DOI: 10.3390/biom11111692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes represents a major health problem, involving a severe imbalance of blood sugar levels, which can disturb the nerves, eyes, kidneys, and other organs. Diabes management involves several synthetic drugs focused on improving insulin sensitivity, increasing insulin production, and decreasing blood glucose levels, but with unclear molecular mechanisms and severe side effects. Natural chemicals extracted from several plants such as Gymnema sylvestre, Momordica charantia or Ophiopogon planiscapus Niger have aroused great interest for their anti-diabetes activity, but also their hypolipidemic and anti-obesity activity. Here, we focused on the anti-diabetic activity of a few natural and synthetic compounds, in correlation with their pharmacokinetic/pharmacodynamic profiles, especially with their blood-brain barrier (BBB) permeability. We reviewed studies that used bioinformatics methods such as predicted BBB, molecular docking, molecular dynamics and quantitative structure-activity relationship (QSAR) to elucidate the proper action mechanisms of antidiabetic compounds. Currently, it is evident that BBB damage plays a significant role in diabetes disorders, but the molecular mechanisms are not clear. Here, we presented the efficacy of natural (gymnemic acids, quercetin, resveratrol) and synthetic (TAK-242, propofol, or APX3330) compounds in reducing diabetes symptoms and improving BBB dysfunctions. Bioinformatics tools can be helpful in the quest for chemical compounds with effective anti-diabetic activity that can enhance the druggability of molecular targets and provide a deeper understanding of diabetes mechanisms.
Collapse
Affiliation(s)
- Ana Maria Udrea
- Laser Department, National Institute for Laser, Plasma and Radiation Physics, 077125 Maurele, Romania; (A.M.U.); (A.D.)
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, University of Bucharest, 1 B. P. Hașdeu St., 50567 Bucharest, Romania;
| | - Gratiela Gradisteanu Pircalabioru
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, University of Bucharest, 1 B. P. Hașdeu St., 50567 Bucharest, Romania;
| | - Anca Andreea Boboc
- “Maria Sklodowska Curie” Emergency Children’s Hospital, 20, Constantin Brancoveanu Bd., 077120 Bucharest, Romania;
- Department of Pediatrics 8, “Carol Davila” University of Medicine and Pharmacy, Eroii Sanitari Bd., 020021 Bucharest, Romania
| | - Catalina Mares
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91–95 Splaiul Independentei, 050095 Bucharest, Romania; (C.M.); (S.A.)
| | - Andra Dinache
- Laser Department, National Institute for Laser, Plasma and Radiation Physics, 077125 Maurele, Romania; (A.M.U.); (A.D.)
| | - Maria Mernea
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91–95 Splaiul Independentei, 050095 Bucharest, Romania; (C.M.); (S.A.)
| | - Speranta Avram
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91–95 Splaiul Independentei, 050095 Bucharest, Romania; (C.M.); (S.A.)
| |
Collapse
|
39
|
Bavnhøj L, Paulsen PA, Flores-Canales JC, Schiøtt B, Pedersen BP. Molecular mechanism of sugar transport in plants unveiled by structures of glucose/H + symporter STP10. NATURE PLANTS 2021; 7:1409-1419. [PMID: 34556835 DOI: 10.1038/s41477-021-00992-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 07/27/2021] [Indexed: 05/28/2023]
Abstract
Sugars are essential sources of energy and carbon and also function as key signalling molecules in plants. Sugar transport proteins (STP) are proton-coupled symporters responsible for uptake of glucose from the apoplast into plant cells. They are integral to organ development in symplastically isolated tissues such as seed, pollen and fruit. Additionally, STPs play a vital role in plant responses to stressors such as dehydration and prevalent fungal infections like rust and mildew. Here we present a structure of Arabidopsis thaliana STP10 in the inward-open conformation at 2.6 Å resolution and a structure of the outward-occluded conformation at improved 1.8 Å resolution, both with glucose and protons bound. The two structures describe key states in the STP transport cycle. Together with molecular dynamics simulations that establish protonation states and biochemical analysis, they pinpoint structural elements, conserved in all STPs, that clarify the basis of proton-to-glucose coupling. These results advance our understanding of monosaccharide uptake, which is essential for plant organ development, and set the stage for bioengineering strategies in crops.
Collapse
Affiliation(s)
- Laust Bavnhøj
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Peter Aasted Paulsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Birgit Schiøtt
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
40
|
Drew D, North RA, Nagarathinam K, Tanabe M. Structures and General Transport Mechanisms by the Major Facilitator Superfamily (MFS). Chem Rev 2021; 121:5289-5335. [PMID: 33886296 PMCID: PMC8154325 DOI: 10.1021/acs.chemrev.0c00983] [Citation(s) in RCA: 221] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/12/2022]
Abstract
The major facilitator superfamily (MFS) is the largest known superfamily of secondary active transporters. MFS transporters are responsible for transporting a broad spectrum of substrates, either down their concentration gradient or uphill using the energy stored in the electrochemical gradients. Over the last 10 years, more than a hundred different MFS transporter structures covering close to 40 members have provided an atomic framework for piecing together the molecular basis of their transport cycles. Here, we summarize the remarkable promiscuity of MFS members in terms of substrate recognition and proton coupling as well as the intricate gating mechanisms undergone in achieving substrate translocation. We outline studies that show how residues far from the substrate binding site can be just as important for fine-tuning substrate recognition and specificity as those residues directly coordinating the substrate, and how a number of MFS transporters have evolved to form unique complexes with chaperone and signaling functions. Through a deeper mechanistic description of glucose (GLUT) transporters and multidrug resistance (MDR) antiporters, we outline novel refinements to the rocker-switch alternating-access model, such as a latch mechanism for proton-coupled monosaccharide transport. We emphasize that a full understanding of transport requires an elucidation of MFS transporter dynamics, energy landscapes, and the determination of how rate transitions are modulated by lipids.
Collapse
Affiliation(s)
- David Drew
- Department
of Biochemistry and Biophysics, Stockholm
University, SE 106 91 Stockholm, Sweden
| | - Rachel A. North
- Department
of Biochemistry and Biophysics, Stockholm
University, SE 106 91 Stockholm, Sweden
| | - Kumar Nagarathinam
- Center
of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Lübeck, D-23538, Lübeck, Germany
| | - Mikio Tanabe
- Structural
Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Oho 1-1, Tsukuba, Ibaraki 305-0801, Japan
| |
Collapse
|
41
|
Tepavčević V. Oligodendroglial Energy Metabolism and (re)Myelination. Life (Basel) 2021; 11:238. [PMID: 33805670 PMCID: PMC7998845 DOI: 10.3390/life11030238] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
Central nervous system (CNS) myelin has a crucial role in accelerating the propagation of action potentials and providing trophic support to the axons. Defective myelination and lack of myelin regeneration following demyelination can both lead to axonal pathology and neurodegeneration. Energy deficit has been evoked as an important contributor to various CNS disorders, including multiple sclerosis (MS). Thus, dysregulation of energy homeostasis in oligodendroglia may be an important contributor to myelin dysfunction and lack of repair observed in the disease. This article will focus on energy metabolism pathways in oligodendroglial cells and highlight differences dependent on the maturation stage of the cell. In addition, it will emphasize that the use of alternative energy sources by oligodendroglia may be required to save glucose for functions that cannot be fulfilled by other metabolites, thus ensuring sufficient energy input for both myelin synthesis and trophic support to the axons. Finally, it will point out that neuropathological findings in a subtype of MS lesions likely reflect defective oligodendroglial energy homeostasis in the disease.
Collapse
Affiliation(s)
- Vanja Tepavčević
- Achucarro Basque Center for Neuroscience, University of the Basque Country, Parque Cientifico de la UPV/EHU, Barrio Sarriena s/n, Edificio Sede, Planta 3, 48940 Leioa, Spain
| |
Collapse
|