1
|
Méndez-Flores OG, Hernández-Kelly LC, Olivares-Bañuelos TN, López-Ramírez G, Ortega A. Brain energetics and glucose transport in metabolic diseases: role in neurodegeneration. Nutr Neurosci 2024; 27:1199-1210. [PMID: 38294500 DOI: 10.1080/1028415x.2024.2306427] [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: 02/01/2024]
Abstract
OBJECTIVES Neurons and glial cells are the main functional and structural elements of the brain, and the former depends on the latter for their nutritional, functional and structural organization, as well as for their energy maintenance. METHODS Glucose is the main metabolic source that fulfills energetic demands, either by direct anaplerosis or through its conversion to metabolic intermediates. Development of some neurodegenerative diseases have been related with modifications in the expression and/or function of glial glucose transporters, which might cause physiological and/or pathological disturbances of brain metabolism. In the present contribution, we summarized the experimental findings that describe the exquisite adjustment in expression and function of glial glucose transporters from physiologic to pathologic metabolism, and its relevance to neurodegenerative diseases. RESULTS A exhaustive literature review was done in order to gain insight into the role of brain energetics in neurodegenerative disease. This study made evident a critical involvement of glucose transporters and thus brain energetics in the development of neurodegenerative diseases. DISCUSSION An exquisite adjustment in the expression and function of glial glucose transporters from physiologic to pathologic metabolism is a biochemical signature of neurodegenerative diseases.
Collapse
Affiliation(s)
- Orquídea G Méndez-Flores
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco (UJAT), Villahermosa, México
| | - Luisa C Hernández-Kelly
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | | | - Gabriel López-Ramírez
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco (UJAT), Villahermosa, México
| | - Arturo Ortega
- Laboratorio de Neurotoxicología, Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
2
|
Barros LF, Schirmeier S, Weber B. The Astrocyte: Metabolic Hub of the Brain. Cold Spring Harb Perspect Biol 2024; 16:a041355. [PMID: 38438188 PMCID: PMC11368191 DOI: 10.1101/cshperspect.a041355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Astrocytic metabolism has taken center stage. Interposed between the neuron and the vasculature, astrocytes exert control over the fluxes of energy and building blocks required for neuronal activity and plasticity. They are also key to local detoxification and waste recycling. Whereas neurons are metabolically rigid, astrocytes can switch between different metabolic profiles according to local demand and the nutritional state of the organism. Their metabolic state even seems to be instructive for peripheral nutrient mobilization and has been implicated in information processing and behavior. Here, we summarize recent progress in our understanding of astrocytic metabolism and its effects on metabolic homeostasis and cognition.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos, Valdivia 5110465, Chile
- Universidad San Sebastián, Facultad de Medicina y Ciencia, Valdivia 5110693, Chile
| | - Stefanie Schirmeier
- Technische Universität Dresden, Department of Biology, 01217 Dresden, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, 8057 Zurich, Switzerland
| |
Collapse
|
3
|
Shaydakov ME, Diaz JA, Eklöf B, Lurie F. Venous valve hypoxia as a possible mechanism of deep vein thrombosis: a scoping review. INT ANGIOL 2024; 43:309-322. [PMID: 38864688 DOI: 10.23736/s0392-9590.24.05170-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
INTRODUCTION The pathogenesis of deep vein thrombosis (DVT) has been explained by an interplay between a changed blood composition, vein wall alteration, and blood flow abnormalities. A comprehensive investigation of these components of DVT pathogenesis has substantially promoted our understanding of thrombogenesis in the venous system. Meanwhile, the process of DVT initiation remains obscure. This systematic review aims to collect, analyze, and synthesize the published evidence to propose hypoxia as a possible trigger of DVT. EVIDENCE ACQUISITION An exhaustive literature search was conducted across multiple electronic databased including PubMed, EMBASE, Scopus, and Web of Science to identify studies pertinent to the research hypothesis. The search was aimed at exploring the connection between hypoxia, reoxygenation, and the initiation of deep vein thrombosis (DVT). The following key words were used: "deep vein thrombosis," "venous thrombosis," "venous thromboembolism," "hypoxia," "reoxygenation," "venous valve," and "venous endothelium." Reviews, case reports, editorials, and letters were excluded. EVIDENCE SYNTHESIS Based on the systematic search outcome, 156 original papers relevant to the issue were selected for detailed review. These studies encompassed a range of experimental and observational clinical research, focusing on various aspects of DVT, including the anatomical, physiological, and cellular bases of the disease. A number of studies suggested limitations in the traditional understanding of Virchow's triad as an acceptable explanation for DVT initiation. Emerging evidence points to more complex interactions and additional factors that may be critical in the early stages of thrombogenesis. The role of venous valves has been recognized but remains underappreciated, with several studies indicating that these sites may act as primary loci for thrombus formation. A collection of studies describes the effects of hypoxia on venous endothelial cells at the cellular and molecular levels. Hypoxia influences several pathways that regulate endothelial cell permeability, inflammatory response, and procoagulation activity, underpinning the endothelial dysfunction noted in DVT. CONCLUSIONS Hypoxia of the venous valve may serve as an independent hypothesis to outline the DVT triggering process. Further research projects in this field may discover new molecular pathways responsible for the disease and suggest new therapeutic targets.
Collapse
Affiliation(s)
- Maxim E Shaydakov
- Division of Vascular Surgery, University of Pittsburgh Medical Center, Pittsburg, PA, USA -
| | - Jose A Diaz
- Division of Surgical Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Fedor Lurie
- Jobst Vascular Institute, ProMedica Health System, Toledo, OH, USA
| |
Collapse
|
4
|
Lopera-Vásquez R, Uribe-García F, Rondón-Barragán I. Effect of estrous cycle phases on gene expression in bovine oviduct epithelial cells. Vet World 2022; 15:1665-1675. [PMID: 36185535 PMCID: PMC9394134 DOI: 10.14202/vetworld.2022.1665-1675] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/25/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: The oviduct environment is of particular importance because it is the site of fertilization and early embryo development. The oviduct, as a component of the reproductive system, responds to ovarian hormone (estradiol [E2] and progesterone [P4]) stimuli depending on the estrous cycle phase. This study aimed to elucidate the effect of estrous cycle phases (follicular and early and late luteal phases) on gene expression patterns in bovine oviduct epithelial cells (BOECs).
Materials and Methods: Oviducts were obtained from healthy slaughterhouse animals, corresponding to ipsilateral ovaries with dominant follicles or corpus luteum during early and late luteal phases. BOECs were recovered from the isthmus (IST) and ampulla (AMP), and the expression patterns of genes related to cytokinesis and mitosis mechanisms (rho-associated coiled-coil containing protein kinase and cellular communication network factor 2 [CCN2]), growth factors (insulin-like growth factor-binding protein 3, epidermal growth factor receptor [EGFR], vascular endothelial growth factor A, and EGFR), antioxidant mechanisms (glutathione peroxidase 4 [GPX4]), apoptosis (B-cell lymphoma 2), complement component (C3), energy metabolism (aldose reductase gene family 1-member b1 [AKRIB1] and solute carrier family 2), hormone receptors (estrogen receptor 1 and luteinizing hormone/choriogonadotropin receptor), and specific glycoproteins (oviductal glycoprotein 1) were analyzed.
Results: High P4 levels (late luteal phase) affected the expression of important genes related to antioxidant mechanisms (GPX4), energy metabolism (AKRIB1), growth factors (IGBP3 and EGFR), and cell growth regulation (CCN2) in the AMP. Low P4 levels (early luteal phase) affected the expression of AKR1B1, IGBP3, and CCN2. In addition, estrogen likely had an effect on OVPGP expression in the cattle oviduct.
Conclusion: Differential gene expression patterns of BOECs in the AMP during the luteal phase (antioxidant mechanisms, energy metabolism, growth factors, and immunological regulators) and in the IST during the follicular phase (glycoproteins) may influence their renewal and population proportions, modulating the oviduct environment as well as gamete and embryo physiology.
Collapse
Affiliation(s)
- Ricaurte Lopera-Vásquez
- Impronta Research Group, Faculty of Veterinary Medicine and Zootechnics. Universidad Cooperativa de Colombia, Ibagué-Tolima, Colombia
| | - Fabián Uribe-García
- Research Group in Immunobiology and Pathogenesis, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Santa Helena Highs, Postal Code 730006299, Ibagué-Tolima, Colombia
| | - Iang Rondón-Barragán
- Research Group in Immunobiology and Pathogenesis, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Santa Helena Highs, Postal Code 730006299, Ibagué-Tolima, Colombia
| |
Collapse
|
5
|
Brain Glucose Transporters: Role in Pathogenesis and Potential Targets for the Treatment of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22158142. [PMID: 34360906 PMCID: PMC8348194 DOI: 10.3390/ijms22158142] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/06/2021] [Accepted: 07/28/2021] [Indexed: 12/16/2022] Open
Abstract
The most common cause of dementia, especially in elderly people, is Alzheimer’s disease (AD), with aging as its main risk factor. AD is a multifactorial neurodegenerative disease. There are several factors increasing the risk of AD development. One of the main features of Alzheimer’s disease is impairment of brain energy. Hypometabolism caused by decreased glucose uptake is observed in specific areas of the AD-affected brain. Therefore, glucose hypometabolism and energy deficit are hallmarks of AD. There are several hypotheses that explain the role of glucose hypometabolism in AD, but data available on this subject are poor. Reduced transport of glucose into neurons may be related to decreased expression of glucose transporters in neurons and glia. On the other hand, glucose transporters may play a role as potential targets for the treatment of AD. Compounds such as antidiabetic drugs, agonists of SGLT1, insulin, siRNA and liposomes are suggested as therapeutics. Nevertheless, the suggested targets of therapy need further investigations.
Collapse
|
6
|
Hertenstein H, McMullen E, Weiler A, Volkenhoff A, Becker HM, Schirmeier S. Starvation-induced regulation of carbohydrate transport at the blood-brain barrier is TGF-β-signaling dependent. eLife 2021; 10:e62503. [PMID: 34032568 PMCID: PMC8149124 DOI: 10.7554/elife.62503] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
During hunger or malnutrition, animals prioritize alimentation of the brain over other organs to ensure its function and, thus, their survival. This protection, also-called brain sparing, is described from Drosophila to humans. However, little is known about the molecular mechanisms adapting carbohydrate transport. Here, we used Drosophila genetics to unravel the mechanisms operating at the blood-brain barrier (BBB) under nutrient restriction. During starvation, expression of the carbohydrate transporter Tret1-1 is increased to provide more efficient carbohydrate uptake. Two mechanisms are responsible for this increase. Similar to the regulation of mammalian GLUT4, Rab-dependent intracellular shuttling is needed for Tret1-1 integration into the plasma membrane; even though Tret1-1 regulation is independent of insulin signaling. In addition, starvation induces transcriptional upregulation that is controlled by TGF-β signaling. Considering TGF-β-dependent regulation of the glucose transporter GLUT1 in murine chondrocytes, our study reveals an evolutionarily conserved regulatory paradigm adapting the expression of sugar transporters at the BBB.
Collapse
Affiliation(s)
- Helen Hertenstein
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Ellen McMullen
- Institut für Neuro- und Verhaltensbiologie, WWU MünsterMünsterGermany
| | - Astrid Weiler
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Anne Volkenhoff
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Holger M Becker
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
- Division of General Zoology, Department of Biology, University of KaiserslauternKaiserslauternGermany
| | - Stefanie Schirmeier
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| |
Collapse
|
7
|
McMullen E, Weiler A, Becker HM, Schirmeier S. Plasticity of Carbohydrate Transport at the Blood-Brain Barrier. Front Behav Neurosci 2021; 14:612430. [PMID: 33551766 PMCID: PMC7863721 DOI: 10.3389/fnbeh.2020.612430] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Neuronal function is highly energy demanding, requiring efficient transport of nutrients into the central nervous system (CNS). Simultaneously the brain must be protected from the influx of unwanted solutes. Most of the energy is supplied from dietary sugars, delivered from circulation via the blood-brain barrier (BBB). Therefore, selective transporters are required to shuttle metabolites into the nervous system where they can be utilized. The Drosophila BBB is formed by perineural and subperineurial glial cells, which effectively separate the brain from the surrounding hemolymph, maintaining a constant microenvironment. We identified two previously unknown BBB transporters, MFS3 (Major Facilitator Superfamily Transporter 3), located in the perineurial glial cells, and Pippin, found in both the perineurial and subperineurial glial cells. Both transporters facilitate uptake of circulating trehalose and glucose into the BBB-forming glial cells. RNA interference-mediated knockdown of these transporters leads to pupal lethality. However, null mutants reach adulthood, although they do show reduced lifespan and activity. Here, we report that both carbohydrate transport efficiency and resulting lethality found upon loss of MFS3 or Pippin are rescued via compensatory upregulation of Tret1-1, another BBB carbohydrate transporter, in Mfs3 and pippin null mutants, while RNAi-mediated knockdown is not compensated for. This means that the compensatory mechanisms in place upon mRNA degradation following RNA interference can be vastly different from those resulting from a null mutation.
Collapse
Affiliation(s)
- Ellen McMullen
- Department of Biology, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Astrid Weiler
- Department of Biology, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Holger M. Becker
- Division of General Zoology, Department of Biology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Stefanie Schirmeier
- Department of Biology, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
8
|
Głuchowska K, Pliszka M, Szablewski L. Expression of glucose transporters in human neurodegenerative diseases. Biochem Biophys Res Commun 2021; 540:8-15. [PMID: 33429199 DOI: 10.1016/j.bbrc.2020.12.067] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
The central nervous system (CNS) plays an important role in the human body. It is involved in the receive, store and participation in information retrieval. It can use several substrates as a source of energy, however, the main source of energy is glucose. Cells of the central nervous system need a continuous supply of energy, therefore, transport of glucose into these cells is very important. There are three distinct families of glucose transporters: sodium-independent glucose transporters (GLUTs), sodium-dependent glucose cotransporters (SGLTs), and uniporter, SWEET protein. In the human brain only GLUTs and SGLTs were detected. In neurodegenerative diseases was observed hypometabolism of glucose due to decreased expression of glucose transporters, in particular GLUT1 and GLUT3. On the other hand, animal studies revealed, that increased levels of these glucose transporters, due to for example by the increased copy number of SLC2A genes, may have a beneficial effect and may be a targeted therapy in the treatment of patients with AD, HD and PD.
Collapse
Affiliation(s)
- Kinga Głuchowska
- Medical University of Warsaw, Chair and Department of General Biology and Parasitology, 5 Chalubinskiego Str., 02-004 Warsaw, Poland.
| | - Monika Pliszka
- Medical University of Warsaw, Chair and Department of General Biology and Parasitology, 5 Chalubinskiego Str., 02-004 Warsaw, Poland.
| | - Leszek Szablewski
- Medical University of Warsaw, Chair and Department of General Biology and Parasitology, 5 Chalubinskiego Str., 02-004 Warsaw, Poland.
| |
Collapse
|
9
|
Espinoza L, Boychuk CR. Diabetes, and its treatment, as an effector of autonomic nervous system circuits and its functions. Curr Opin Pharmacol 2020; 54:18-26. [PMID: 32721846 DOI: 10.1016/j.coph.2020.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022]
Abstract
Diabetes increases the risk of cardiovascular complications, including heart failure, hypertension, and stroke. There is a strong involvement of autonomic dysfunction in individuals with diabetes that exhibit clinical manifestations of cardiovascular diseases (CVD). Still, the mechanisms by which diabetes and its treatments alter autonomic function and subsequently affect cardiovascular complications remain elusive. For this reason, understanding the brainstem circuits involved in sensing metabolic state(s) and enacting autonomic control of the cardiovascular system are important to develop more comprehensive therapies for individuals with diabetes at increased risk for CVD. We review how autonomic nervous system circuits change during these disease states and discuss their potential role in current pharmacotherapies that target diabetic states. Overall, this review proposes that the brainstem circuits provide an integrative sensorimotor network capable of responding to metabolic cues to regulate cardiovascular function and this network is modified by, and in turn affects, diabetes-induced CVD and its treatment.
Collapse
Affiliation(s)
- Liliana Espinoza
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, United States
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, United States.
| |
Collapse
|
10
|
Leão LL, Tangen G, Barca ML, Engedal K, Santos SHS, Machado FSM, de Paula AMB, Monteiro-Junior RS. Does hyperglycemia downregulate glucose transporters in the brain? Med Hypotheses 2020; 139:109614. [PMID: 32087490 DOI: 10.1016/j.mehy.2020.109614] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Diabetes is a metabolic condition associated with hyperglycemia manifested by the elevation of blood glucose levels occurring when the pancreas decreases or stops the production of insulin, in case of insulin resistance or both. The current literature supports that insulin resistance may be responsible for the memory decline associated with diabetes. Glucose transporters (GLUTs) are a family of proteins involved in glucose transport across biological membranes. GLUT-1 and GLUT-3 are involved in glucose delivery to the brain. Evidence suggests that both transporters are downregulated in chronic peripheral hyperglycemia. Here we show the mechanisms of glucose transport and its influence on cognitive function, including a hypothesis of how peripheral hyperglycemia related genes network interactions may lead to glucose transporters downregulation and its possible consequences.
Collapse
Affiliation(s)
- Luana Lemos Leão
- Post-graduate Program of Health Sciences, State University of Montes Claros, Montes Claros, Minas Gerais, Brazil
| | - Gro Tangen
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
| | - Maria Lage Barca
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
| | - Knut Engedal
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
| | - Sérgio Henrique S Santos
- Post-graduate Program of Health Sciences, State University of Montes Claros, Montes Claros, Minas Gerais, Brazil; Institute of Agricultural Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Frederico Sander M Machado
- Post-graduate Program of Health Sciences, State University of Montes Claros, Montes Claros, Minas Gerais, Brazil
| | - Alfredo Maurício B de Paula
- Post-graduate Program of Health Sciences, State University of Montes Claros, Montes Claros, Minas Gerais, Brazil
| | - Renato Sobral Monteiro-Junior
- Post-graduate Program of Health Sciences, State University of Montes Claros, Montes Claros, Minas Gerais, Brazil; Post-Graduate Program of Medicine (Neurology/Neuroscience), Federal Fluminense University, Niterói, Rio de Janeiro, Brazil; Neuroscience of Exercise Institute, Aroldo Tourinho Hospital, Montes Claros, MG, Brazil.
| |
Collapse
|
11
|
Abstract
Diabetes is a global epidemic and a leading cause of death with more than 422 million patients worldwide out of whom around 392 million alone suffer from type 2 diabetes (T2D). Sodium-glucose cotransporter 2 inhibitors (SGLT2i) are novel and effective drugs in managing glycemia of T2D patients. These inhibitors gained recent clinical and basic research attention due to their clinically observed cardiovascular protective effects. Although interest in the study of various SGLT isoforms and the effect of their inhibition on cardiovascular function extends over the past 20 years, an explanation of the effects observed clinically based on available experimental data is not forthcoming. The remarkable reduction in cardiovascular (CV) mortality (38%), major CV events (14%), hospitalization for heart failure (35%), and death from any cause (32%) observed over a period of 2.6 years in patients with T2D and high CV risk in the EMPA-REG OUTCOME trial involving the SGLT2 inhibitor empagliflozin (Empa) have raised the possibility that potential novel, more specific mechanisms of SGLT2 inhibition synergize with the known modest systemic improvements, such as glycemic, body weight, diuresis, and blood pressure control. Multiple studies investigated the direct impact of SGLT2i on the cardiovascular system with limited findings and the pathophysiological role of SGLTs in the heart. The direct impact of SGLT2i on cardiac homeostasis remains controversial, especially that SGLT1 isoform is the only form expressed in the capillaries and myocardium of human and rodent hearts. The direct impact of SGLT2i on the cardiovascular system along with potential lines of future research is summarized in this review.
Collapse
|
12
|
Young TL, Zychowski KE, Denson JL, Campen MJ. Blood-brain barrier at the interface of air pollution-associated neurotoxicity and neuroinflammation. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Rojas-Gutierrez E, Muñoz-Arenas G, Treviño S, Espinosa B, Chavez R, Rojas K, Flores G, Díaz A, Guevara J. Alzheimer's disease and metabolic syndrome: A link from oxidative stress and inflammation to neurodegeneration. Synapse 2017. [PMID: 28650104 DOI: 10.1002/syn.21990] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and one of the most important causes of morbidity and mortality among the aging population. AD diagnosis is made post-mortem, and the two pathologic hallmarks, particularly evident in the end stages of the illness, are amyloid plaques and neurofibrillary tangles. Currently, there is no curative treatment for AD. Additionally, there is a strong relation between oxidative stress, metabolic syndrome, and AD. The high levels of circulating lipids and glucose imbalances amplify lipid peroxidation that gradually diminishes the antioxidant systems, causing high levels of oxidative metabolism that affects cell structure, leading to neuronal damage. Accumulating evidence suggests that AD is closely related to a dysfunction of both insulin signaling and glucose metabolism in the brain, leading to an insulin-resistant brain state. Four drugs are currently used for this pathology: Three FDA-approved cholinesterase inhibitors and one NMDA receptor antagonist. However, wide varieties of antioxidants are promissory to delay or prevent the symptoms of AD and may help in treating the disease. Therefore, therapeutic efforts to achieve attenuation of oxidative stress could be beneficial in AD treatment, attenuating Aβ-induced neurotoxicity and improve neurological outcomes in AD. The term inflammaging characterizes a widely accepted paradigm that aging is accompanied by a low-grade chronic up-regulation of certain pro-inflammatory responses in the absence of overt infection, and is a highly significant risk factor for both morbidity and mortality in the elderly.
Collapse
Affiliation(s)
- Eduardo Rojas-Gutierrez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Guadalupe Muñoz-Arenas
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Samuel Treviño
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Blanca Espinosa
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias-INER, Ciudad de México, Mexico
| | - Raúl Chavez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Karla Rojas
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Alfonso Díaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Jorge Guevara
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
14
|
Weiler A, Volkenhoff A, Hertenstein H, Schirmeier S. Metabolite transport across the mammalian and insect brain diffusion barriers. Neurobiol Dis 2017; 107:15-31. [PMID: 28237316 DOI: 10.1016/j.nbd.2017.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 01/02/2017] [Accepted: 02/20/2017] [Indexed: 12/31/2022] Open
Abstract
The nervous system in higher vertebrates is separated from the circulation by a layer of specialized endothelial cells. It protects the sensitive neurons from harmful blood-derived substances, high and fluctuating ion concentrations, xenobiotics or even pathogens. To this end, the brain endothelial cells and their interlinking tight junctions build an efficient diffusion barrier. A structurally analogous diffusion barrier exists in insects, where glial cell layers separate the hemolymph from the neural cells. Both types of diffusion barriers, of course, also prevent influx of metabolites from the circulation. Because neuronal function consumes vast amounts of energy and necessitates influx of diverse substrates and metabolites, tightly regulated transport systems must ensure a constant metabolite supply. Here, we review the current knowledge about transport systems that carry key metabolites, amino acids, lipids and carbohydrates into the vertebrate and Drosophila brain and how this transport is regulated. Blood-brain and hemolymph-brain transport functions are conserved and we can thus use a simple, genetically accessible model system to learn more about features and dynamics of metabolite transport into the brain.
Collapse
Affiliation(s)
- Astrid Weiler
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Anne Volkenhoff
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Helen Hertenstein
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Stefanie Schirmeier
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany.
| |
Collapse
|
15
|
Szablewski L. Glucose Transporters in Brain: In Health and in Alzheimer’s Disease. J Alzheimers Dis 2016; 55:1307-1320. [DOI: 10.3233/jad-160841] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
16
|
Glucose Transporters at the Blood-Brain Barrier: Function, Regulation and Gateways for Drug Delivery. Mol Neurobiol 2016; 54:1046-1077. [PMID: 26801191 DOI: 10.1007/s12035-015-9672-6] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/17/2015] [Indexed: 12/31/2022]
Abstract
Glucose transporters (GLUTs) at the blood-brain barrier maintain the continuous high glucose and energy demands of the brain. They also act as therapeutic targets and provide routes of entry for drug delivery to the brain and central nervous system for treatment of neurological and neurovascular conditions and brain tumours. This article first describes the distribution, function and regulation of glucose transporters at the blood-brain barrier, the major ones being the sodium-independent facilitative transporters GLUT1 and GLUT3. Other GLUTs and sodium-dependent transporters (SGLTs) have also been identified at lower levels and under various physiological conditions. It then considers the effects on glucose transporter expression and distribution of hypoglycemia and hyperglycemia associated with diabetes and oxygen/glucose deprivation associated with cerebral ischemia. A reduction in glucose transporters at the blood-brain barrier that occurs before the onset of the main pathophysiological changes and symptoms of Alzheimer's disease is a potential causative effect in the vascular hypothesis of the disease. Mutations in glucose transporters, notably those identified in GLUT1 deficiency syndrome, and some recreational drug compounds also alter the expression and/or activity of glucose transporters at the blood-brain barrier. Approaches for drug delivery across the blood-brain barrier include the pro-drug strategy whereby drug molecules are conjugated to glucose transporter substrates or encapsulated in nano-enabled delivery systems (e.g. liposomes, micelles, nanoparticles) that are functionalised to target glucose transporters. Finally, the continuous development of blood-brain barrier in vitro models is important for studying glucose transporter function, effects of disease conditions and interactions with drugs and xenobiotics.
Collapse
|
17
|
Zhang S, Song XY, Xia CY, Ai QD, Chen J, Chu SF, He WB, Chen NH. Effects of cerebral glucose levels in infarct areas on stroke injury mediated by blood glucose changes. RSC Adv 2016. [DOI: 10.1039/c6ra19715a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
18
|
Sajja RK, Prasad S, Cucullo L. Impact of altered glycaemia on blood-brain barrier endothelium: an in vitro study using the hCMEC/D3 cell line. Fluids Barriers CNS 2014; 11:8. [PMID: 24708805 PMCID: PMC3985548 DOI: 10.1186/2045-8118-11-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/17/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cerebrovascular complications involving endothelial dysfunction at the blood-brain barrier (BBB) are central to the pathogenesis of diabetes-related CNS disorders. However, clinical and experimental studies have reported contrasting evidence in relation to the effects of hyperglycemia on BBB permeability and function. Similarly the effect of hypoglycemia on BBB integrity is not well understood. Therefore, we assessed the differential impact of hypo and hyperglycemic conditions on BBB integrity and endothelial function in vitro using hCMEC/D3, a well characterized human brain microvascular endothelial cell line. METHODS Parallel monolayers of hCMEC/D3 were exposed to normal, hypo- or hyperglycemic media, containing 5.5, 2.2 or 35 mM D-glucose, respectively. Following 3-24h exposure, the expression and distribution of BBB tight junction (ZO-1 and claudin-5) adherence junction (VE-cadherin) proteins, and glucose transporters as well as inflammatory (VCAM-1) and oxidative stress (Nrf-2) markers were analyzed by immunofluorescence and western blotting. Endothelial release of growth factors and pro-inflammatory cytokines were determined by ELISA. Further, the impact of altered glycemia on BBB permeability was assessed in hCMEC/D3 - astrocyte co-cultures on Transwell supports using fluorescent dextrans (4-70 kDa). RESULTS Compared to controls, exposure to hypoglycemia (3 and 24h) down-regulated the expression of claudin-5 and disrupted the ZO-1 localization at cell-cell contacts, while hyperglycemia marginally reduced claudin-5 expression without affecting ZO-1 distribution. Permeability to dextrans (4-10 kDa) and VEGF release at 24h were significantly increased by hypo- and hyperglycemia, although 70 kDa dextran permeability was increased only under hypoglycemic conditions. The expression of SGLT-1 was up-regulated at 24h hypoglycemic exposure while only a modest increase of GLUT-1 expression was observed. In addition, the expression of Nrf-2 and release of interleukin-6 and PDGF-BB, were down-regulated by hypoglycemia (but not hyperglycemia), while both conditions induced a marginal and transient increase in VCAM-1 expression from 3 to 24h, including a significant increase in VE-cadherin expression at 3 h following hyperglycemia. CONCLUSIONS In summary, our findings demonstrate a potential impairment of BBB integrity and function by hypo or hyperglycemia, through altered expression/distribution of TJ proteins and nutrient transporters. In addition, hypoglycemic exposure severely affects the expression of oxidative and inflammatory stress markers of BBB endothelium.
Collapse
Affiliation(s)
| | | | - Luca Cucullo
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S, Coulter Street, Amarillo, TX 79106, USA.
| |
Collapse
|
19
|
Tumor Cell Uptake of 99mTc-Labeled 1-Thio-β-d-Glucose and 5-Thio-d-Glucose in Comparison with 2-Deoxy-2-[18 F]Fluoro-d-Glucose In Vitro: Kinetics, Dependencies, Blockage and Cell Compartment of Accumulation. Mol Imaging Biol 2013; 16:189-98. [DOI: 10.1007/s11307-013-0690-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
20
|
Seto SW, Au ALS, Poon CCW, Zhang Q, Li RWS, Yeung JHK, Kong SK, Ngai SM, Wan S, Ho HP, Lee SMY, Hoi MPM, Chan SW, Leung GPH, Kwan YW. Acute simvastatin inhibits K ATP channels of porcine coronary artery myocytes. PLoS One 2013; 8:e66404. [PMID: 23799098 PMCID: PMC3684588 DOI: 10.1371/journal.pone.0066404] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/06/2013] [Indexed: 01/19/2023] Open
Abstract
Background Statins (3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors) consumption provides beneficial effects on cardiovascular systems. However, effects of statins on vascular KATP channel gatings are unknown. Methods Pig left anterior descending coronary artery and human left internal mammary artery were isolated and endothelium-denuded for tension measurements and Western immunoblots. Enzymatically-dissociated/cultured arterial myocytes were used for patch-clamp electrophysiological studies and for [Ca2+]i, [ATP]i and [glucose]o uptake measurements. Results The cromakalim (10 nM to 10 µM)- and pinacidil (10 nM to 10 µM)-induced concentration-dependent relaxation of porcine coronary artery was inhibited by simvastatin (3 and 10 µM). Simvastatin (1, 3 and 10 µM) suppressed (in okadaic acid (10 nM)-sensitive manner) cromakalim (10 µM)- and pinacidil (10 µM)-mediated opening of whole-cell KATP channels of arterial myocytes. Simvastatin (10 µM) and AICAR (1 mM) elicited a time-dependent, compound C (1 µM)-sensitive [3H]-2-deoxy-glucose uptake and an increase in [ATP]i levels. A time (2–30 min)- and concentration (0.1–10 µM)-dependent increase by simvastatin of p-AMPKα-Thr172 and p-PP2A-Tyr307 expression was observed. The enhanced p-AMPKα-Thr172 expression was inhibited by compound C, ryanodine (100 µM) and KN93 (10 µM). Simvastatin-induced p-PP2A-Tyr307 expression was suppressed by okadaic acid, compound C, ryanodine, KN93, phloridzin (1 mM), ouabain (10 µM), and in [glucose]o-free or [Na+]o-free conditions. Conclusions Simvastatin causes ryanodine-sensitive Ca2+ release which is important for AMPKα-Thr172 phosphorylation via Ca2+/CaMK II. AMPKα-Thr172 phosphorylation causes [glucose]o uptake (and an [ATP]i increase), closure of KATP channels, and phosphorylation of AMPKα-Thr172 and PP2A-Tyr307 resulted. Phosphorylation of PP2A-Tyr307 occurs at a site downstream of AMPKα-Thr172 phosphorylation.
Collapse
Affiliation(s)
- Sai Wang Seto
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, School of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Alice Lai Shan Au
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Christina Chui Wa Poon
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Qian Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Rachel Wai Sum Li
- Department of Pharmacology and Pharmacy, Faculty of Medicine, The University of Hong Kong, Hong Kong, PR of China
| | - John Hok Keung Yeung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Siu Kai Kong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Sai Ming Ngai
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Song Wan
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Ho Pui Ho
- Department of Electronic Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Simon Ming Yuen Lee
- Institute of Chinese Medical Sciences, the University of Macau, Macau, PR of China
| | - Maggie Pui Man Hoi
- Institute of Chinese Medical Sciences, the University of Macau, Macau, PR of China
| | - Shun Wan Chan
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, PR of China
- * E-mail: (YWK); (GPHL); (SWC)
| | - George Pak Heng Leung
- Department of Pharmacology and Pharmacy, Faculty of Medicine, The University of Hong Kong, Hong Kong, PR of China
- * E-mail: (YWK); (GPHL); (SWC)
| | - Yiu Wa Kwan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
- * E-mail: (YWK); (GPHL); (SWC)
| |
Collapse
|
21
|
Yu AS, Hirayama BA, Timbol G, Liu J, Diez-Sampedro A, Kepe V, Satyamurthy N, Huang SC, Wright EM, Barrio JR. Regional distribution of SGLT activity in rat brain in vivo. Am J Physiol Cell Physiol 2012; 304:C240-7. [PMID: 23151803 DOI: 10.1152/ajpcell.00317.2012] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Na(+)-glucose cotransporter (SGLT) mRNAs have been detected in many organs of the body, but, apart from kidney and intestine, transporter expression, localization, and functional activity, as well as physiological significance, remain elusive. Using a SGLT-specific molecular imaging probe, α-methyl-4-deoxy-4-[(18)F]fluoro-D-glucopyranoside (Me-4-FDG) with ex vivo autoradiography and immunohistochemistry, we mapped in vivo the regional distribution of functional SGLTs in rat brain. Since Me-4-FDG is not a substrate for GLUT1 at the blood-brain barrier (BBB), in vivo delivery of the probe into the brain was achieved after opening of the BBB by an established procedure, osmotic shock. Ex vivo autoradiography showed that Me-4-FDG accumulated in regions of the cerebellum, hippocampus, frontal cortex, caudate nucleus, putamen, amygdala, parietal cortex, and paraventricular nucleus of the hypothalamus. Little or no Me-4-FDG accumulated in the brain stem. The regional accumulation of Me-4-FDG overlapped the distribution of SGLT1 protein detected by immunohistochemistry. In summary, after the BBB is opened, the specific substrate for SGLTs, Me-4-FDG, enters the brain and accumulates in selected regions shown to express SGLT1 protein. This localization and the sensitivity of these neurons to anoxia prompt the speculation that SGLTs may play an essential role in glucose utilization under stress such as ischemia. The expression of SGLTs in the brain raises questions about the potential effects of SGLT inhibitors under development for the treatment of diabetes.
Collapse
Affiliation(s)
- Amy S Yu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
The occurrence of altered brain glucose metabolism has long been suggested in both diabetes and Alzheimer’s diseases. However, the preceding mechanism to altered glucose metabolism has not been well understood. Glucose enters the brain via glucose transporters primarily present at the blood-brain barrier. Any changes in glucose transporter function and expression dramatically affects brain glucose homeostasis and function. In the brains of both diabetic and Alzheimer’s disease patients, changes in glucose transporter function and expression have been observed, but a possible link between the altered glucose transporter function and disease progress is missing. Future recognition of the role of new glucose transporter isoforms in the brain may provide a better understanding of brain glucose metabolism in normal and disease states. Elucidation of clinical pathological mechanisms related to glucose transport and metabolism may provide common links to the etiology of these two diseases. Considering these facts, in this review we provide a current understanding of the vital roles of a variety of glucose transporters in the normal, diabetic and Alzheimer’s disease brain.
Collapse
Affiliation(s)
- Kaushik Shah
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX 79106, USA.
| | | | | |
Collapse
|
23
|
Kanwal A, Singh SP, Grover P, Banerjee SK. Development of a cell-based nonradioactive glucose uptake assay system for SGLT1 and SGLT2. Anal Biochem 2012; 429:70-5. [DOI: 10.1016/j.ab.2012.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/23/2012] [Accepted: 07/03/2012] [Indexed: 11/25/2022]
|
24
|
Bormans G, De Groot T, Van Oosterwijck G, Mortelmans L, Verbruggen A. Synthesis and biological evaluation of [11C]Methyl-D-glucoside, a tracer for the sodium-glucose cotransporter (SGLT). J Labelled Comp Radiopharm 2012. [DOI: 10.1002/jlcr.25804401101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
25
|
Targeting vascular amyloid in arterioles of Alzheimer disease transgenic mice with amyloid β protein antibody-coated nanoparticles. J Neuropathol Exp Neurol 2011; 70:653-61. [PMID: 21760540 DOI: 10.1097/nen.0b013e318225038c] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The relevance of cerebral amyloid angiopathy (CAA) to the pathogenesis of Alzheimer disease (AD) and dementia in general emphasizes the importance of developing novel targeting approaches for detecting and treating cerebrovascular amyloid (CVA) deposits. We developed a nanoparticle-based technology that uses a monoclonal antibody against fibrillar human amyloid-β42 that is surface coated onto a functionalized phospholipid monolayer. We demonstrate that this conjugated nanoparticle binds to CVA deposits in arterioles of AD transgenic mice (Tg2576) after infusion into the external carotid artery using 3 different approaches. The first 2 approaches use a blood vessel enrichment of homogenized brain and a leptomeningeal vessel preparation from thin tangential brain slices from the surface of the cerebral cortex. Targeting of CVA by the antibody-coated nanoparticle was visualized using fluorescent lissamine rhodamine-labeled phospholipids in the nanoparticles, which were compared with fluorescent staining of the endothelial cells and amyloid deposits using confocal laser scanning microscopy. The third approach used high-field strength magnetic resonance imaging of antibody-coated iron oxide nanoparticles after infusion into the external carotid artery. Dark foci of contrast enhancement in cortical arterioles were observed in T2*-weighted images of ex vivo AD mouse brains that correlated histologically with CVA deposits. The targeting ability of these nanoparticles to CVA provides opportunities for the prevention and treatment of CAA.
Collapse
|
26
|
Gaudreault N, Scriven DRL, Moore EDW. Asymmetric Subcellular Distribution of Glucose Transporters in the Endothelium of Small Contractile Arteries. ACTA ACUST UNITED AC 2009; 13:317-24. [PMID: 17090404 DOI: 10.1080/10623320600972085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The authors have recently reported the presence and asymmetric distribution of the glucose transporters GLUT-1 to -5 and SGLT-1 in the endothelium of rat coronary artery (Gaudreault et al. 2004, Diabetologica, 47, 2081-2092). In the present study the authors investigate and compare the presence and subcellular distribution of the classic glucose transporter isoforms in endothelial cells of cerebral, renal, and mesenteric arteries. The GLUTs and SGLT-1 were examined with immunohistochemistry and wide-field fluorescence microscopy coupled to deconvolution in en face preparation of intact artery. We identified GLUT-1 to -5 and SGLT-1 in the endothelial cells of all three vascular beds. The relative level of expression for each isoform was found comparable amongst arteries. Clusters of the glucose transporter isoforms were found at a high density in proximity to the cell-to-cell junctions. In addition, a consistent asymmetric distribution of GLUT-1 to -5 was found, predominantly located on the abluminal side of the endothelium in all three vascular beds examined (ranging from 68% to 91%, p<.05). The authors conclude that the expression and subcellular distribution of glucose transporters are similar in endothelial cells from vascular beds of comparable diameter and suggest that their subcellular organization may facilitate transendothelial transport of glucose in small contractile arteries.
Collapse
Affiliation(s)
- N Gaudreault
- Department of Physiology, University of British Columbia, Vancouver, Canada
| | | | | |
Collapse
|
27
|
Vemula S, Roder KE, Yang T, Bhat GJ, Thekkumkara TJ, Abbruscato TJ. A functional role for sodium-dependent glucose transport across the blood-brain barrier during oxygen glucose deprivation. J Pharmacol Exp Ther 2008; 328:487-95. [PMID: 18981287 DOI: 10.1124/jpet.108.146589] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the current study, we determined the functional significance of sodium-dependent/-independent glucose transporters at the neurovasculature during oxygen glucose deprivation (OGD). Confluent brain endothelial cells cocultured with astrocytes were exposed to varying degrees of in vitro stroke conditions. Glucose transporter (GLUT) 1 and sodium glucose cotransporter (SGLT) activity were investigated by luminal membrane uptake and transport studies using [(3)H]D-glucose and also by [(14)C]alpha-methyl D-glucopyranoside (AMG), a specific, nonmetabolized substrate of SGLT. In vivo middle cerebral artery occlusion experiments were tested to determine whether blood-brain barrier (BBB) SGLT activity was induced during ischemia. Increases in luminal D-glucose and AMG uptake and transport were observed with in vitro stroke conditions. Specific inhibitor experiments suggest a combined role for both SGLT and GLUT1 at the BBB during OGD. A time-dependent increase in the uptake of AMG was also seen in mice exposed to permanent focal ischemia, and this increase was sensitive to the SGLT inhibitor, phlorizin. Infarct and edema ratio during ischemia were significantly decreased by the inhibition of this transporter. These results show that both GLUT1 and SGLT play a role at the BBB in the blood-to-brain transport of glucose during ischemic conditions, and inhibition of SGLT during stroke has the potential to improve stroke outcome. Pharmacological modulation of this novel BBB transporter could prove to be a brain vascular target in stroke.
Collapse
Affiliation(s)
- Sharanya Vemula
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79016, USA
| | | | | | | | | | | |
Collapse
|
28
|
Role of beta-adrenoceptors in memory consolidation: beta3-adrenoceptors act on glucose uptake and beta2-adrenoceptors on glycogenolysis. Neuropsychopharmacology 2008; 33:2384-97. [PMID: 18046311 DOI: 10.1038/sj.npp.1301629] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Noradrenaline, acting via beta(2)- and beta(3)-adrenoceptors (AR), enhances memory formation in single trial-discriminated avoidance learning in day-old chicks by mechanisms involving changes in metabolism of glucose and/or glycogen. Earlier studies of memory consolidation in chicks implicated beta(3)- rather than beta(2)-ARs in enhancement of memory consolidation by glucose, but did not elucidate whether stimulation of glucose uptake or of glycolysis was responsible. This study examines the role of glucose transport in memory formation using central injection of the nonselective facilitative glucose transporter (GLUT) inhibitor cytochalasin B, the endothelial/astrocytic GLUT-1 inhibitor phloretin and the Na(+)/energy-dependent endothelial glucose transporter (SGLT) inhibitor phlorizin. Cytochalasin B inhibited memory when injected into the mesopallium (avian cortex) either close to or between 25 and 45 min after training, whereas phloretin and phlorizin only inhibited memory at 30 min. This suggested that astrocytic/endothelial (GLUT-1) transport is critical at the time of consolidation, whereas a different transporter, probably the neuronal glucose transporter (GLUT-3), is important at the time of training. Inhibition of glucose transport by cytochalasin B, phloretin, or phlorizin also interfered with beta(3)-AR-mediated memory enhancement 20 min posttraining, whereas inhibition of glycogenolysis interfered with beta(2)-AR agonist enhancement of memory. We conclude that in astrocytes (1) activities of both GLUT-1 and SGLT are essential for memory consolidation 30 min posttraining; (2) neuronal GLUT-3 is essential at the time of training; and (3) beta(2)- and beta(3)-ARs consolidate memory by different mechanisms; beta(3)-ARs stimulate central glucose transport, whereas beta(2)-ARs stimulate central glycogenolysis.
Collapse
|
29
|
Yang T, Roder KE, Abbruscato TJ. Evaluation of bEnd5 cell line as an in vitro model for the blood-brain barrier under normal and hypoxic/aglycemic conditions. J Pharm Sci 2008; 96:3196-213. [PMID: 17828743 DOI: 10.1002/jps.21002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The purpose of the study was to assess the suitability of the mouse endothelial cell line bEnd5 as a blood-brain barrier (BBB) model under normal or pathologic (stroke) conditions. In comparison to the well-established bovine brain endothelial cell (BBMEC) model, cultured bEnd5 monolayers reached a maximal transendothelial electrical resistance (TEER) of 121 Omega cm(2) on day 7, and possessed oval and spindle shape morphology. Structurally, confluent monolayers of bEnd5 cells and BBMECs exhibit peripheral band staining of the tight junction protein ZO-1 and occludin. Both bEnd5 and BBMECs express important tight junctional proteins, ZO-1, occludin and claudin-1, as well as the transporters P-glycoprotein (P-gp), NKCC, GLUT1, and most PKC isoforms. Marker permeability experiments suggest that bEnd5 cells form a tight barrier that compares to well-established in vitro BBB models, such as the BBMEC. After short durations of hypoxia/aglycemia (H/A), hyperpermeability was seen in the bEnd5 endothelial monolayer compared to later time periods for BBMECs, suggesting that bEnd5 cells are more sensitive to hypoxia/algycemia treatment than BBMECs. Taken together, bEnd5 cell culture model may provide a useful in vitro model of the BBB for drug delivery studies and modeling pathological states such as oxygen glucose deprivation associated with stroke.
Collapse
Affiliation(s)
- Tianzhi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University, Health Sciences Center, 1300 Coulter Drive, Amarillo, TX 79106, USA
| | | | | |
Collapse
|
30
|
Gaudreault N, Scriven DRL, Laher I, Moore EDW. Subcellular characterization of glucose uptake in coronary endothelial cells. Microvasc Res 2008; 75:73-82. [PMID: 17531273 DOI: 10.1016/j.mvr.2007.04.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Revised: 03/06/2007] [Accepted: 04/02/2007] [Indexed: 11/18/2022]
Abstract
Despite all the evidence linking glucose toxicity to an increased risk of cardiovascular diseases, very little is known about the regulation of glucose uptake in endothelial cells. We have previously reported an asymmetric distribution of the GLUTs (1-5) and SGLT-1 in en face preparations of rat coronary artery endothelia [Gaudreault N., Scriven D.R., Moore E.D., 2004. Characterisation of glucose transporters in the intact coronary artery endothelium in rats: GLUT-2 upregulated by long-term hyperglycaemia. Diabetologia 47(12),2081-2092]. We assessed this time, through immunocytochemistry and wide field fluorescence microscopy coupled to deconvolution, the presence and subcellular distribution of glucose transporters in cultures of human coronary artery endothelial cells (HCAECs). HCAECs express GLUT-1 to 5 and SGLT-1, but their subcellular distribution lacks the luminal/abluminal asymmetry and the proximity to cell-to-cell junctions observed in intact endothelium. To determine the impact of the transporters' distribution on intracellular glucose accumulation, a fluorescent glucose analog (2-NBDG) was used in conjunction with confocal microscopy to monitor uptake in individual cells; the arteries were mounted in an arteriograph chamber with physiological flow rates. The uptake in both preparations was inhibited by cytochalasin-B and d-glucose and stimulated by insulin, but the distribution of the incorporated 2-NBDG mirrored that of the transporters. In HCAEC it was distributed throughout the cell and in the intact arterial endothelium it was restricted to the narrow cytosolic volume adjacent to the cell-to-cell junctions. We suggest that the latter subcellular organization and compartmentalization may facilitate transendothelial transport of glucose in intact coronary artery.
Collapse
Affiliation(s)
- N Gaudreault
- Department of Cellular and Physiological Sciences, University of British Columbia, 2146 Health Sciences Mall, Vancouver, B.C., Canada
| | | | | | | |
Collapse
|
31
|
Chan CY, Guggenheim JA, To CH. Is active glucose transport present in bovine ciliary body epithelium? Am J Physiol Cell Physiol 2006; 292:C1087-93. [PMID: 17020938 DOI: 10.1152/ajpcell.00048.2006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperglycemia is a major risk factor for diabetic cataract formation. Effective regulation of glucose transport by the ciliary body epithelium (CBE) is pivotal to normal glycemic control in the anterior eye, which in turn affects the glucose level of the crystalline lens. The present study aimed to characterize the glucose transport mechanisms across the bovine blood-aqueous barrier (BAB) represented by the CBE. With an Ussing-type chamber, the glucose transport kinetics were measured and characterized in the presence and absence of various glucose transporter inhibitors. The saturation characteristics of the CBE to glucose were estimated from an Eadie-Hofstee plot. The mRNA expression of glucose transporters in specific regions of the bovine CBE was assessed using RT-PCR. The trans-CBE glucose flux was found to be sensitive to the glucose transporter inhibitors cytochalasin B, phloretin, and phlorizin. The transport system had a kinetic constant of 5.3 mM and a maximum velocity of 349.5 nmol.h(-1).cm(-2). Gene expression for GLUT1, GLUT3, GLUT4, GLUT5, and SGLT2 was observed in both the pars plana and pars plicata regions of the bovine CBE. This study demonstrates that glucose transport across the bovine CBE is primarily passive in nature. However, the novel findings of 1) the presence of a phlorizin-sensitive glucose flux and 2) gene expression for SGLT2 mean that a potential role for active glucose transport cannot be ruled out. The elucidation of the exact function of SGLT2 in the bovine CBE may shed important light on the glucose transport and physiology of the BAB and inform future studies of glycemic control in relation to diabetic cataract formation.
Collapse
Affiliation(s)
- Chu Yan Chan
- School of Optometry and Vision Sciences, Cardiff University, Wales, UK
| | | | | |
Collapse
|
32
|
Qutub AA, Hunt CA. Glucose transport to the brain: a systems model. ACTA ACUST UNITED AC 2005; 49:595-617. [PMID: 16269321 DOI: 10.1016/j.brainresrev.2005.03.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2004] [Revised: 03/02/2005] [Accepted: 03/09/2005] [Indexed: 02/07/2023]
Abstract
Glucose transport to the brain involves sophisticated interactions of solutes, transporters, enzymes, and cell signaling processes, within an intricate spatial architecture. The dynamics of the transport are influenced by the adaptive nature of the blood-brain barrier (BBB), the semi-impermeable membranes of brain capillaries. As both the gate and the gatekeeper between blood-borne nutrients and brain tissue, the BBB helps govern brain homeostasis. Glucose in the blood must cross the BBB's luminal and abluminal membranes to reach neural tissue. A robust representation of the glucose transport mechanism can highlight a target for brain therapeutic intervention, help characterize mechanisms behind several disease phenotypes, or suggest a new delivery route for drugs. The challenge for researchers is understanding the relationships between influential physiological variables in vivo, and using that knowledge to predict how alterations or interventions affect glucose transport. This paper reviews factors influencing glucose transport and approaches to representing blood-to-brain glucose transport including in vitro, in vivo, and kinetic models. Applications for different models are highlighted, while their limitations in answering arising questions about the human in vivo BBB lead to a discussion of an alternate approach. A developing complex systems simulation is introduced, initiating a single platform to represent the dynamics of glucose transport across the adapting human blood-brain barrier.
Collapse
Affiliation(s)
- Amina A Qutub
- Joint Graduate Group in Bioengineering, University of California, Berkeley and San Francisco, USA.
| | | |
Collapse
|
33
|
Gaudreault N, Scriven DRL, Moore EDW. Characterisation of glucose transporters in the intact coronary artery endothelium in rats: GLUT-2 upregulated by long-term hyperglycaemia. Diabetologia 2004; 47:2081-92. [PMID: 15662550 DOI: 10.1007/s00125-004-1583-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2004] [Accepted: 07/18/2004] [Indexed: 01/22/2023]
Abstract
AIMS/HYPOTHESIS We have examined the effects of streptozotocin-induced type 1 diabetes on the expression and subcellular distribution of the classic sugar transporters (GLUT-1 to 5 and sodium-dependent glucose transporter-1 [SGLT-1]) in the endothelial cells of an en face preparation of septal coronary artery from Wistar rats. METHODS The presence of the GLUT isoforms and SGLT-1 in the endothelial cell layer was determined by immunohistochemistry using wide-field fluorescence microscopy coupled to deconvolution, and was quantified by digital image analysis. RESULTS We found that all of the transporters were expressed within these cells and that all except SGLT-1 were preferentially located on the abluminal side. The heaviest labelling was adjacent to the cell-to-cell junctions where the luminal and abluminal membranes are in close proximity, which may reflect a spatial organisation specialised for vectorial glucose transport across the thinnest part of the cytoplasm. Long-term hyperglycaemia, induced by streptozotocin, significantly downregulated GLUT-1, 3, 4 and 5 and dramatically upregulated GLUT-2, leaving SGLT-1 unchanged. CONCLUSIONS/INTERPRETATION We conclude that the high susceptibility of endothelial cells to glucose toxicity may be the result of the subcellular organisation of their GLUTs and the increased expression of GLUT-2.
Collapse
Affiliation(s)
- N Gaudreault
- Department of Physiology, University of British Columbia, 2146 Health Sciences Mall, Vancouver, B.C., V6T 1Z3, Canada
| | | | | |
Collapse
|
34
|
Taubert D, Rosenkranz A, Berkels R, Roesen R, Schömig E. Acute effects of glucose and insulin on vascular endothelium. Diabetologia 2004; 47:2059-71. [PMID: 15662548 DOI: 10.1007/s00125-004-1586-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2004] [Accepted: 07/22/2004] [Indexed: 10/26/2022]
Abstract
AIMS/HYPOTHESIS Chronic exposure to high concentrations of glucose has consistently been demonstrated to impair endothelium-dependent, nitric oxide (NO)-mediated vasodilation. In contrast, several clinical investigations have reported that acute exposure to high glucose, alone or in combination with insulin, triggers vasodilation. The aim of this study was to examine whether elevated glucose itself stimulates endothelial NO formation or enhances insulin-mediated endothelial NO release. METHODS We measured NO release and vessel tone ex vivo in porcine coronary conduit arteries (PCAs). Intracellular Ca(2+) was monitored in porcine aortic endothelial cells (PAECs) by fura-2 fluorescence. Expression of the Na(+)/glucose cotransporter-1 (SGLT-1) was assayed in PAECs and PCA endothelium by RT-PCR. RESULTS Stimulation of PCAs with D: -glucose, but not the osmotic control L: -glucose, induced a transient increase in NO release (EC(50) approximately 10 mmol/l), mediated by a rise in intracellular Ca(2+) levels due to an influx from the extracellular space. This effect was abolished by inhibitors of the plasmalemmal Na(+)/Ca(2+) exchanger (dichlorobenzamil) and the SGLT-1 (phlorizin), which was found to be expressed in aortic and coronary endothelium. Alone, D: -glucose did not relax PCA, but did augment the effect of insulin on NO release and vasodilation. CONCLUSIONS/INTERPRETATION An increased supply of extracellular D: -glucose appears to enhance the activity of the endothelial isoform of nitric oxide synthase by increasing intracellular Na(+) concentrations via SGLT-1, which in turn stimulates an extracellular Ca(2+) influx through the Na(+)/Ca(2+) exchanger. This mechanism may be responsible for glucose-enhanced, insulin-dependent increases in tissue perfusion (including coronary blood-flow), thus accelerating glucose extraction from the blood circulation to limit the adverse vascular effects of prolonged hyperglycaemia.
Collapse
Affiliation(s)
- D Taubert
- Department of Pharmacology, Medical Hospital of the University of Cologne, Gleueler Str. 24, 50931, Cologne, Germany.
| | | | | | | | | |
Collapse
|
35
|
Romanelli L, Amico MC, Mattioli F, Memoli A, Savini G, Muller A. Effects of insulin-induced acute hypoglycemia and normoglycemic hyperinsulinemia on the retinal uptake and ocular metabolism of glucose in rabbits. Metabolism 2004; 53:1274-83. [PMID: 15375782 DOI: 10.1016/j.metabol.2004.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glucose is the principal metabolic substrate for the retina in mammals, being essential for maintaining the functional activity of the retina; it can be supplied to the tissue by both vitreous humor and blood. Yet, the impact of hypoglycemia on retinal glucose metabolism has been poorly investigated. We have therefore studied the effects of acute insulin-induced hypoglycemia on the glucose uptake and metabolism in the retina, by analyzing the hypoglycemia-induced changes in the ocular distribution and metabolic fate of [3H]-2-deoxy-D-glucose (2-DG) and [14C]-D-glucose, both injected in the vitreous body. Rabbits were rendered hypoglycemic by subcutaneous injection of insulin (0.8 and 1.2 IU/kg). Insulin-induced hypoglycemia increased both retinal [3H]-radioactivity levels and retina to vitreous humor ratio of [3H]-radioactivity levels ([3H]-[R/VH]). Radio-chromatography showed that hypoglycemia did not induce any change in the retinal conversion of 2-DG to 2-DG-6-phosphate, but increased the conversion of [14C]-D-glucose to [14C]-lactate. Normoglycemic hyperinsulinemia caused no change in either retinal [3H]-radioactivity levels or [3H]-[R/VH] while decreasing retinal [14C]-radioactivity levels and retina to vitreous ratios of 14C-radioactivity levels. These results indicate that acute hypoglycemia increases the uptake rate of glucose by the retina and suggest that normoglycemic hyperinsulinemia may decrease retinal lactate, possibly stimulating its removal from the retina.
Collapse
Affiliation(s)
- Luca Romanelli
- Department of Pharmacology of Natural Substances, Faculty of Pharmacy, University of Rome La Sapienza, Italy
| | | | | | | | | | | |
Collapse
|
36
|
Li H, Gu Y, Zhang Y, Lucas MJ, Wang Y. High glucose levels down-regulate glucose transporter expression that correlates with increased oxidative stress in placental trophoblast cells in vitro. ACTA ACUST UNITED AC 2004; 11:75-81. [PMID: 14980308 DOI: 10.1016/j.jsgi.2003.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To study glucose transporter expression and oxidative stress in placental trophoblasts under hyperglycemic conditions in vitro. METHODS Trophoblasts were isolated from term normal human placentas and incubated with Dulbecco's modified eagle medium containing 1000, 2500, and 4500 mg/L glucose for 3 days. At the end of incubation, culture medium was collected. Trophoblast RNA was extracted and mRNA expression of glucose transporters was determined by RNase protection assay. Messenger RNA expression for copper-zinc-superoxide dismutase (CuZn-SOD) was determined by real-time polymerase chain reaction. Lipid peroxide production was determined by measuring malondialdehyde concentration in the culture supernatant. Protein expression of sodium-glucose transporter 2 (SGLT-2) was determined by Western blot analysis. RESULTS Messenger RNA expression for glucose transporter 1 (GLUT1) and SGLT-2 were reduced in trophoblast cells incubated with 4500 mg/L glucose compared with those incubated with 1000 and 2000 mg/L glucose. mRNA expression of CuZn-SOD was also decreased in trophoblasts incubated with 4500 mg/L glucose. Malondialdehyde production was significantly increased by trophoblasts incubated with 4500 mg/L glucose compared with those by trophoblasts incubated with 1000 and 2000 mg/L glucose (4.69 +/- 0.60 versus 2.10 +/- 0.29 and 2.89 +/- 0.47 nmol/mg protein; P < .01, respectively). CONCLUSIONS Down-regulation of gene expression of glucose transporters correlates with increased lipid peroxide production and decreased superoxide dismutase expression in placental trophoblasts cultured under hyperglycemic conditions.
Collapse
Affiliation(s)
- Hui Li
- Department of Obstetrics and Gynecology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | | | | | | | |
Collapse
|
37
|
Elfeber K, Stümpel F, Gorboulev V, Mattig S, Deussen A, Kaissling B, Koepsell H. Na(+)-D-glucose cotransporter in muscle capillaries increases glucose permeability. Biochem Biophys Res Commun 2004; 314:301-5. [PMID: 14733905 DOI: 10.1016/j.bbrc.2003.12.090] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
By immunohistochemistry, we demonstrated the localization of the Na(+)-D-glucose cotransporter SGLT1 in capillaries of rat heart and skeletal muscle, but not in capillaries of small intestine and submandibular gland. mRNA of SGLT1 was identified in skeletal muscle and primary cultured coronary endothelial cells. The functional relevance of SGLT1 for glucose transport across capillary walls in muscle was tested by measuring the extraction of D-glucose from the perfusate during non-recirculating perfusion of isolated rat hindlimbs. In this model, D-glucose extraction from the perfusate is increased by insulin which accelerates D-glucose uptake into myocytes by increasing the concentration of glucose transporter GLUT4 in the plasma membrane. The insulin-induced increase of D-glucose extraction from the perfusate was abolished after blocking SGLT1 with the specific inhibitor phlorizin. The data show that SGLT1 in capillaries of skeletal muscle is required for the action of insulin on D-glucose supply of myocytes.
Collapse
Affiliation(s)
- Katrin Elfeber
- Institut für Anatomie und Zellbiologie der Bayerischen Julius-Maximilians-Universität, Koellikerstrasse 6, 97070 Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
38
|
Elfeber K, Köhler A, Lutzenburg M, Osswald C, Galla HJ, Witte OW, Koepsell H. Localization of the Na+-D-glucose cotransporter SGLT1 in the blood-brain barrier. Histochem Cell Biol 2004; 121:201-7. [PMID: 14986005 DOI: 10.1007/s00418-004-0633-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2004] [Indexed: 10/26/2022]
Abstract
Immunoreactivity of the Na+-D-glucose cotransporter SGLT1 was demonstrated in intracerebral capillaries of rat and pig. Immunostaining suggested that SGLT1 is located in the luminal membrane of the endothelial cells and in intracellular vesicles. Using in situ hybridization, SGLT1 mRNA was not detectable in intracerebral capillaries of non-treated or sham-operated Wistar rats. However, 1 day after a transient occlusion of the right middle cerebral artery, SGLT1 mRNA was detected in capillaries of both brain hemispheres. Expression of SGLT1 was also demonstrated in primary cultures of capillary endothelial cells from pig using polymerase chain reaction after reverse transcription and western blotting. The data suggest that SGLT1 participates in transport of D-glucose across the blood-brain barrier and is upregulated after brain ischemia and reperfusion.
Collapse
Affiliation(s)
- Katrin Elfeber
- Institute of Anatomy and Cell Biology, University Würzburg, Koellikerstrasse 6, 97070 Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Nakamura K, Takayama K, Nagai T, Maitani Y. Regional intestinal absorption of FITC-dextran 4,400 with nanoparticles based on beta-sitosterol beta-D-glucoside in rats. J Pharm Sci 2003; 92:311-8. [PMID: 12532381 DOI: 10.1002/jps.10292] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nanoparticles (NP) are potential carriers for drug delivery to the targeted intestine. NP based on beta-sitosterol beta-D-glucoside (Sit-G) enhanced the colon-specific absorption of FITC-dextran 4,400 (FD-4), because the concentration-dependent increase of bioavailability appeared in only the colon. In a permeation study, the absorption enhancement in the colon was suppressed in the following conditions: (1) the addition of Sit-G NP to serosa; (2) a permeation study at 4 degrees C; (3) the addition of endocytosis inhibitor, cytochalasin B. NP based on sitosterol, the aglycon of Sit-G, did not increase the FD-4 colonic permeation. The addition of Sit-G NP to the mucosal side induced a decrease of transepithelial resistance (TEER), but this phenomenon was suppressed by an inhibitor of Na(+)-dependent specific glucose transporter, phrolidzin, which did not affect FD-4 permeation. These findings suggested that absorption enhancement by Sit-G NP may not be due to opening of a tight junction, but might be related to endocytosis via glucose residue of Sit-G.
Collapse
Affiliation(s)
- Koji Nakamura
- Department of Pharmaceutics, Hoshi University, Ebara 2-4-41, Shinagawa, Tokyo 142-8501, Japan
| | | | | | | |
Collapse
|
40
|
Mann GE, Yudilevich DL, Sobrevia L. Regulation of amino acid and glucose transporters in endothelial and smooth muscle cells. Physiol Rev 2003; 83:183-252. [PMID: 12506130 DOI: 10.1152/physrev.00022.2002] [Citation(s) in RCA: 319] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
While transport processes for amino acids and glucose have long been known to be expressed in the luminal and abluminal membranes of the endothelium comprising the blood-brain and blood-retinal barriers, it is only within the last decades that endothelial and smooth muscle cells derived from peripheral vascular beds have been recognized to rapidly transport and metabolize these nutrients. This review focuses principally on the mechanisms regulating amino acid and glucose transporters in vascular endothelial cells, although we also summarize recent advances in the understanding of the mechanisms controlling membrane transport activity and expression in vascular smooth muscle cells. We compare the specificity, ionic dependence, and kinetic properties of amino acid and glucose transport systems identified in endothelial cells derived from cerebral, retinal, and peripheral vascular beds and review the regulation of transport by vasoactive agonists, nitric oxide (NO), substrate deprivation, hypoxia, hyperglycemia, diabetes, insulin, steroid hormones, and development. In view of the importance of NO as a modulator of vascular tone under basal conditions and in disease and chronic inflammation, we critically review the evidence that transport of L-arginine and glucose in endothelial and smooth muscle cells is modulated by bacterial endotoxin, proinflammatory cytokines, and atherogenic lipids. The recent colocalization of the cationic amino acid transporter CAT-1 (system y(+)), nitric oxide synthase (eNOS), and caveolin-1 in endothelial plasmalemmal caveolae provides a novel mechanism for the regulation of NO production by L-arginine delivery and circulating hormones such insulin and 17beta-estradiol.
Collapse
Affiliation(s)
- Giovanni E Mann
- Centre for Cardiovascular Biology and Medicine, Guy's, King's, and St. Thomas' School of Biomedical Sciences, King's College London, London, United Kingdom.
| | | | | |
Collapse
|
41
|
Aussedat B, Dupire-Angel M, Gifford R, Klein JC, Wilson GS, Reach G. Interstitial glucose concentration and glycemia: implications for continuous subcutaneous glucose monitoring. Am J Physiol Endocrinol Metab 2000; 278:E716-28. [PMID: 10751207 DOI: 10.1152/ajpendo.2000.278.4.e716] [Citation(s) in RCA: 140] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The changes in plasma glucose concentration and in interstitial glucose concentration, determined with a miniaturized subcutaneous glucose sensor, were investigated in anesthetized nondiabetic rats. Interstitial glucose was estimated through two different calibration procedures. First, after a glucose load, the magnitude of the increase in interstitial glucose, estimated through a one-point calibration procedure, was 70% of that in plasma glucose. We propose that this is due to the effect of endogenous insulin on peripheral glucose uptake. Second, during the spontaneous secondary decrease in plasma glucose after the glucose load, interstitial glucose decreased faster than plasma glucose, which may also be due to the effect of insulin on peripheral glucose uptake. Third, during insulin-induced hypoglycemia, the decrease in interstitial glucose was less marked than that of plasma glucose, suggesting that hypoglycemia suppressed transfer of glucose into the interstitial tissue; subsequently, interstitial glucose remained lower than plasma glucose during its return to basal value, suggesting that the stimulatory effect of insulin on peripheral glucose uptake was protracted. If these observations obtained in rats are relevant to human physiology, such discrepancies between plasma and interstitial glucose concentration may have major implications for the use of a subcutaneous glucose sensor in continuous blood glucose monitoring in diabetic patients.
Collapse
Affiliation(s)
- B Aussedat
- Department of Diabetology, Institut National de la Santé et de la Recherche Médicale U341, Hôtel-Dieu, 75004 Paris, France
| | | | | | | | | | | |
Collapse
|