1
|
González-Johnson L, Fariña A, Farías G, Zomosa G, Pinilla-González V, Rojas-Solé C. Exploring Neuroprotection against Radiation-Induced Brain Injury: A Review of Key Compounds. NEUROSCI 2024; 5:462-484. [PMID: 39484304 PMCID: PMC11503407 DOI: 10.3390/neurosci5040034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/03/2024] Open
Abstract
Brain radiation is a crucial tool in neuro-oncology for enhancing local tumor control, but it can lead to mild-to-profound and progressive impairments in cognitive function. Radiation-induced brain injury is a significant adverse effect of radiotherapy for cranioencephalic tumors, primarily caused by indirect cellular damage through the formation of free radicals. This results in late neurotoxicity manifesting as cognitive impairment due to free radical production. The aim of this review is to highlight the role of different substances, such as drugs used in the clinical setting and antioxidants such as ascorbate, in reducing the neurotoxicity associated with radiation-induced brain injury. Currently, there is mainly preclinical and clinical evidence supporting the benefit of these interventions, representing a cost-effective and straightforward neuroprotective strategy.
Collapse
Affiliation(s)
- Lucas González-Johnson
- Faculty of Medicine, Universidad de Chile, Santiago 8330111, Chile; (G.F.); (V.P.-G.); (C.R.-S.)
- University of Chile Clinical Hospital, Santiago 8380453, Chile;
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, Universidad de Chile, Santiago 8330111, Chile
| | - Ariel Fariña
- Fundación Arturo López Pérez, Santiago 7500921, Chile;
- Faculty of Medicine, Universidad de los Andes, Santiago 12455, Chile
| | - Gonzalo Farías
- Faculty of Medicine, Universidad de Chile, Santiago 8330111, Chile; (G.F.); (V.P.-G.); (C.R.-S.)
- University of Chile Clinical Hospital, Santiago 8380453, Chile;
| | - Gustavo Zomosa
- University of Chile Clinical Hospital, Santiago 8380453, Chile;
| | - Víctor Pinilla-González
- Faculty of Medicine, Universidad de Chile, Santiago 8330111, Chile; (G.F.); (V.P.-G.); (C.R.-S.)
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8330111, Chile
| | - Catalina Rojas-Solé
- Faculty of Medicine, Universidad de Chile, Santiago 8330111, Chile; (G.F.); (V.P.-G.); (C.R.-S.)
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8330111, Chile
| |
Collapse
|
2
|
Burgess ER, Praditi C, Phillips E, Vissers MCM, Robinson BA, Dachs GU, Wiggins GAR. Role of Sodium-Dependent Vitamin C Transporter-2 and Ascorbate in Regulating the Hypoxic Pathway in Cultured Glioblastoma Cells. J Cell Biochem 2024:e30658. [PMID: 39382087 DOI: 10.1002/jcb.30658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/21/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
The most common and aggressive brain cancer, glioblastoma, is characterized by hypoxia and poor survival. The pro-tumour transcription factor, hypoxia-inducible factor (HIF), is regulated via HIF-hydroxylases that require ascorbate as cofactor. Decreased HIF-hydroxylase activity triggers the hypoxic pathway driving cancer progression. Tissue ascorbate accumulates via the sodium-dependent vitamin C transporter-2 (SVCT2). We hypothesize that glioblastoma cells rely on SVCT2 for ascorbate accumulation, and that knockout of this transporter would disrupt the regulation of the hypoxic pathway by ascorbate. Ascorbate uptake was measured in glioblastoma cell lines (U87MG, U251MG, T98G) by high-performance liquid chromatography. CRISPR/Cas9 was used to knockout SVCT2. Cells were treated with cobalt chloride, desferrioxamine or 5% oxygen, with/without ascorbate, and key hypoxic pathway proteins were measured using Western blot analysis. Ascorbate uptake was cell line dependent, ranging from 1.7 to 11.0 nmol/106 cells. SVCT2-knockout cells accumulated 90%-95% less intracellular ascorbate than parental cells. The hypoxic pathway was induced by all three stimuli, and ascorbate reduced this induction. In the SVCT2-knockout cells, ascorbate had limited effect on the hypoxic pathway. This study verifies that intracellular ascorbate is required to suppress the hypoxic pathway. As patient survival is related to an activated hypoxic pathway, increasing intra-tumoral ascorbate may be of clinical interest.
Collapse
Affiliation(s)
- Eleanor R Burgess
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunology (MI3), Heidelberg University, Medical Faculty Mannheim, Mannheim, Germany
| | - Citra Praditi
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
- Mātai Hāora, Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Margreet C M Vissers
- Mātai Hāora, Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Bridget A Robinson
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
- Canterbury Regional Cancer and Haematology Service, Te Whatu Ora, Waitaha/Canterbury, Christchurch, New Zealand
| | - Gabi U Dachs
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - George A R Wiggins
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
3
|
Saldivia N, Salazar K, Cifuentes M, Espinoza F, Harrison FE, Nualart F. Ascorbic acid and its transporter SVCT2, affect radial glia cells differentiation in postnatal stages. Glia 2024; 72:708-727. [PMID: 38180226 DOI: 10.1002/glia.24498] [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: 03/03/2023] [Revised: 12/03/2023] [Accepted: 12/10/2023] [Indexed: 01/06/2024]
Abstract
Radial glia (RG) cells generate neurons and glial cells that make up the cerebral cortex. Both in rodents and humans, these stem cells remain for a specific time after birth, named late radial glia (lRG). The knowledge of lRG and molecules that may be involved in their differentiation is based on very limited data. We analyzed whether ascorbic acid (AA) and its transporter SVCT2, are involved in lRG cells differentiation. We demonstrated that lRG cells are highly present between the first and fourth postnatal days. Anatomical characterization of lRG cells, revealed that lRG cells maintained their bipolar morphology and stem-like character. When lRG cells were labeled with adenovirus-eGFP at 1 postnatal day, we detected that some cells display an obvious migratory neuronal phenotype, suggesting that lRG cells continue generating neurons postnatally. Moreover, we demonstrated that SVCT2 was apically polarized in lRG cells. In vitro studies using the transgenic mice SVCT2+/- and SVCT2tg (SVCT2-overexpressing mouse), showed that decreased SVCT2 levels led to accelerated differentiation into astrocytes, whereas both AA treatment and elevated SVCT2 expression maintain the lRG cells in an undifferentiated state. In vivo overexpression of SVCT2 in lRG cells generated cells with a rounded morphology that were migratory and positive for proliferation and neuronal markers. We also examined mediators that can be involved in AA/SVCT2-modulated signaling pathways, determining that GSK3-β through AKT, mTORC2, and PDK1 is active in brains with high levels of SVCT2/AA. Our data provide new insights into the role of AA and SVCT2 in late RG cells.
Collapse
Affiliation(s)
- Natalia Saldivia
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy, CMA BIO BIO, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Katterine Salazar
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy, CMA BIO BIO, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Manuel Cifuentes
- Department of Cell Biology, Genetics and Physiology, Universidad de Málaga, IBIMA, Málaga, Spain
| | - Francisca Espinoza
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy, CMA BIO BIO, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Fiona E Harrison
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, USA
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy, CMA BIO BIO, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
4
|
Long Y, Yi C, Wu R, Zhang Y, Zhang B, Shi X, Zhang X, Zha Z. Biodistribution and radiation dosimetry in cancer patients of the ascorbic acid analogue 6-Deoxy-6-[ 18F] fluoro-L-ascorbic acid PET imaging: first-in-human study. Eur J Nucl Med Mol Imaging 2023; 50:3072-3083. [PMID: 37191679 DOI: 10.1007/s00259-023-06262-9] [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: 12/25/2022] [Accepted: 05/06/2023] [Indexed: 05/17/2023]
Abstract
PURPOSE Clinical studies on the use of ascorbic acid (AA) have become a hot spot in cancer research. There remains an unmet need to assess AA utilization in normal tissues and tumors. 6-Deoxy-6-[18F]fluoro-L-ascorbic acid ([18F]DFA) displayed distinctive tumor localization and similar distribution as AA in mice. In this study, to evaluate the distribution, tumor detecting ability and radiation dosimetry of [18F]DFA in humans, we performed the first-in-human PET imaging study. METHODS Six patients with a variety of cancers underwent whole-body PET/CT scans after injection of 313-634 MBq of [18F]DFA. Five sequential dynamic emission scans in each patient were acquired at 5-60 min. Regions of interest (ROI) were delineated along the edge of the source-organ and tumor on the transverse PET slice. Tumor-to-background ratio (TBR) was obtained using the tumor SUVmax to background SUVmean. Organ residence times were calculated via time-activity curves, and human absorbed doses were estimated from organ residence time using the medical internal radiation dosimetry method. RESULTS [18F]DFA was well tolerated in all subjects without serious adverse event. The high uptake was found in the liver, adrenal glands, kidneys, choroid plexus, and pituitary gland. [18F]DFA accumulated in tumor rapidly and the TBR increased over time. The average SUVmax of [18F]DFA in tumor lesions was 6.94 ± 3.92 (range 1.62-22.85, median 5.94). The organs with the highest absorbed doses were the liver, spleen, adrenal glands, and kidneys. The mean effective dose was estimated to be 1.68 ± 0.36 E-02 mSv/MBq. CONCLUSIONS [18F]DFA is safe to be used in humans. It showed a similar distribution pattern as AA, and displayed high uptake and retention in tumors with appropriate kinetics. [18F]DFA might be a promising radiopharmaceutical in identifying tumors with high affinity for SVCT2 and monitoring AA distribution in both normal tissues and tumors. TRIAL REGISTRATION Chinese Clinical Trial Registry; Registered Number: ChiCTR2200057842 (registered 19 March 2022).
Collapse
Affiliation(s)
- Yali Long
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Chang Yi
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Renbo Wu
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Yuying Zhang
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Bing Zhang
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Xinchong Shi
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Xiangsong Zhang
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Zhihao Zha
- Department of Nuclear Medicine, the First Affiliated Hospital of Sun Yat-sen University, 58# Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| |
Collapse
|
5
|
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
|
6
|
Morris-Blanco KC, Chokkalla AK, Kim T, Bhatula S, Bertogliat MJ, Gaillard AB, Vemuganti R. High-Dose Vitamin C Prevents Secondary Brain Damage After Stroke via Epigenetic Reprogramming of Neuroprotective Genes. Transl Stroke Res 2022; 13:1017-1036. [PMID: 35306630 PMCID: PMC9485293 DOI: 10.1007/s12975-022-01007-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/27/2022] [Accepted: 03/14/2022] [Indexed: 12/27/2022]
Abstract
Vitamin C has recently been identified as an epigenetic regulator by activating ten-eleven translocases (TETs), enzymes involved in generating DNA hydroxymethylcytosine (5hmC). Currently, we investigated whether high-dose vitamin C promotes neuroprotection through epigenetic modulation of 5hmC, if there are sex-specific differences in outcome, and the therapeutic potential of vitamin C in stroke-related comorbidities in adult mice. Post-stroke treatment with ascorbate (reduced form), but not dehydroascorbate (oxidized form), increased TET3 activity and 5hmC levels and reduced infarct following focal ischemia. Hydroxymethylation DNA immunoprecipitation sequencing showed that ascorbate increased 5hmC across the genome and specifically in promoters of several stroke pathophysiology-related genes, particularly anti-inflammatory genes. Ascorbate also decreased markers of oxidative stress, mitochondrial fragmentation, and apoptosis in cortical peri-infarct neurons and promoted motor and cognitive functional recovery in both sexes via TET3. Furthermore, post-stroke ascorbate treatment reduced infarct volume and improved motor function recovery in aged, hypertensive and diabetic male and female mice. Delayed ascorbate treatment at 6 h of reperfusion was still effective at reducing infarct volume and motor impairments in adult mice. Collectively, this study shows that post-stroke treatment with high-dose ascorbate protects the brain through epigenetic reprogramming and may function as a robust therapeutic against stroke injury.
Collapse
Affiliation(s)
- Kahlilia C Morris-Blanco
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
- Cellular and Molecular Pathology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - TaeHee Kim
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
| | - Saivenkateshkomal Bhatula
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| | - Mario J Bertogliat
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
| | - Alexis B Gaillard
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin-Madison, Mail code CSC-8660, 600 Highland Ave, Madison, WI, 53792, USA.
- William S. Middleton Veterans Administration Hospital, Madison, WI, USA.
- Cellular and Molecular Pathology Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
7
|
The Membrane Electrical Potential and Intracellular pH as Factors Influencing Intracellular Ascorbate Concentration and Their Role in Cancer Treatment. Cells 2021; 10:cells10112964. [PMID: 34831187 PMCID: PMC8616305 DOI: 10.3390/cells10112964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Ascorbate is an important element of a variety of cellular processes including the control of reactive oxygen species levels. Since reactive oxygen species are implicated as a key factor in tumorigenesis and antitumor therapy, the injection of a large amount of ascorbate is considered beneficial in cancer therapy. Recent studies have shown that ascorbate can cross the plasma membrane through passive diffusion. In contrast to absorption by active transport, which is facilitated by transport proteins (SVCT1 and SVCT2). The passive diffusion of a weak acid across membranes depends on the electrostatic potential and the pH gradients. This has been used to construct a new theoretical model capable of providing steady-state ascorbate concentration in the intracellular space and evaluating the time needed to reach it. The main conclusion of the analysis is that the steady-state intracellular ascorbate concentration weakly depends on its serum concentration but requires days of exposure to saturate. Based on these findings, it can be hypothesized that extended oral ascorbate delivery is possibly more effective than a short intravenous infusion of high ascorbate quantities.
Collapse
|
8
|
Tveden-Nyborg P. Vitamin C Deficiency in the Young Brain-Findings from Experimental Animal Models. Nutrients 2021; 13:1685. [PMID: 34063417 PMCID: PMC8156420 DOI: 10.3390/nu13051685] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022] Open
Abstract
Severe and long-term vitamin C deficiency can lead to fatal scurvy, which is fortunately considered rare today. However, a moderate state of vitamin C (vitC) deficiency (hypovitaminosis C)-defined as a plasma concentration below 23 μM-is estimated to affect up to 10% of the population in the Western world, albeit clinical hallmarks in addition to scurvy have not been linked to vitC deficiency. The brain maintains a high vitC content and uniquely high levels during deficiency, supporting vitC's importance in the brain. Actions include both antioxidant and co-factor functions, rendering vitamin C deficiency likely to affect several targets in the brain, and it could be particularly significant during development where a high cellular metabolism and an immature antioxidant system might increase sensitivity. However, investigations of a non-scorbutic state of vitC deficiency and effects on the developing young brain are scarce. This narrative review provides a comprehensive overview of the complex mechanisms that regulate vitC homeostasis in vivo and in the brain in particular. Functions of vitC in the brain and the potential consequences of deficiency during brain development are highlighted, based primarily on findings from experimental animal models. Perspectives for future investigations of vitC are outlined.
Collapse
Affiliation(s)
- Pernille Tveden-Nyborg
- Section of Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Copenhagen, Denmark
| |
Collapse
|
9
|
Renner O, Burkard M, Michels H, Vollbracht C, Sinnberg T, Venturelli S. Parenteral high‑dose ascorbate - A possible approach for the treatment of glioblastoma (Review). Int J Oncol 2021; 58:35. [PMID: 33955499 PMCID: PMC8104923 DOI: 10.3892/ijo.2021.5215] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/05/2021] [Indexed: 12/14/2022] Open
Abstract
For glioblastoma, the treatment with standard of care therapy comprising resection, radiation, and temozolomide results in overall survival of approximately 14-18 months after initial diagnosis. Even though several new therapy approaches are under investigation, it is difficult to achieve life prolongation and/or improvement of patient's quality of life. The aggressiveness and progression of glioblastoma is initially orchestrated by the biological complexity of its genetic phenotype and ability to respond to cancer therapy via changing its molecular patterns, thereby developing resistance. Recent clinical studies of pharmacological ascorbate have demonstrated its safety and potential efficacy in different cancer entities regarding patient's quality of life and prolongation of survival. In this review article, the actual glioblastoma treatment possibilities are summarized, the evidence for pharmacological ascorbate in glioblastoma treatment is examined and questions are posed to identify current gaps of knowledge regarding accessibility of ascorbate to the tumor area. Experiments with glioblastoma cell lines and tumor xenografts have demonstrated that high-dose ascorbate induces cytotoxicity and oxidative stress largely selectively in malignant cells compared to normal cells suggesting ascorbate as a potential therapeutic agent. Further investigations in larger cohorts and randomized placebo-controlled trials should be performed to confirm these findings as well as to improve delivery strategies to the brain, through the inherent barriers and ultimately to the malignant cells.
Collapse
Affiliation(s)
- Olga Renner
- Department of Nutritional Biochemistry, University of Hohenheim, D‑70599 Stuttgart, Germany
| | - Markus Burkard
- Department of Nutritional Biochemistry, University of Hohenheim, D‑70599 Stuttgart, Germany
| | - Holger Michels
- Pascoe Pharmazeutische Praeparate GmbH, D‑35394 Giessen, Germany
| | | | - Tobias Sinnberg
- Department of Dermatology, University Hospital Tuebingen, D‑72076 Tuebingen, Germany
| | - Sascha Venturelli
- Department of Nutritional Biochemistry, University of Hohenheim, D‑70599 Stuttgart, Germany
| |
Collapse
|
10
|
High-Dose Vitamin C: Preclinical Evidence for Tailoring Treatment in Cancer Patients. Cancers (Basel) 2021; 13:cancers13061428. [PMID: 33804775 PMCID: PMC8003833 DOI: 10.3390/cancers13061428] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/02/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Vitamin C is an indispensable micronutrient in the human diet due to the multiple functions it carries out in the body. Reports of clinical studies have indicated that, when administered at high dosage by the intravenous route, vitamin C may exert beneficial antitumor effects in patients with advanced stage cancers, including those refractory to previous treatment with chemotherapy. The aim of this article is to provide an overview of the current scientific evidence concerning the different mechanisms of action by which high-dose vitamin C may kill tumor cells. A special focus will be given to those mechanisms that provide the rationale basis for tailoring vitamin C treatment according to specific molecular alterations present in the tumor and for the selection of the most appropriate companion drugs. Abstract High-dose vitamin C has been proposed as a potential therapeutic approach for patients with advanced tumors who failed previous treatment with chemotherapy. Due to vitamin C complex pharmacokinetics, only intravenous administration allows reaching sufficiently high plasma concentrations required for most of the antitumor effects observed in preclinical studies (>0.250 mM). Moreover, vitamin C entry into cells is tightly regulated by SVCT and GLUT transporters, and is cell type-dependent. Importantly, besides its well-recognized pro-oxidant effects, vitamin C modulates TET enzymes promoting DNA demethylation and acts as cofactor of HIF hydroxylases, whose activity is required for HIF-1α proteasomal degradation. Furthermore, at pharmacological concentrations lower than those required for its pro-oxidant activity (<1 mM), vitamin C in specific genetic contexts may alter the DNA damage response by increasing 5-hydroxymethylcytosine levels. These more recently described vitamin C mechanisms offer new treatment opportunities for tumors with specific molecular defects (e.g., HIF-1α over-expression or TET2, IDH1/2, and WT1 alterations). Moreover, vitamin C action at DNA levels may provide the rationale basis for combination therapies with PARP inhibitors and hypomethylating agents. This review outlines the pharmacokinetic and pharmacodynamic properties of vitamin C to be taken into account in designing clinical studies that evaluate its potential use as anticancer agent.
Collapse
|
11
|
(Ascorb)ing Pb Neurotoxicity in the Developing Brain. Antioxidants (Basel) 2020; 9:antiox9121311. [PMID: 33371438 PMCID: PMC7767447 DOI: 10.3390/antiox9121311] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Lead (Pb) neurotoxicity is a major concern, particularly in children. Developmental exposure to Pb can alter neurodevelopmental trajectory and has permanent neuropathological consequences, including an increased vulnerability to further stressors. Ascorbic acid is among most researched antioxidant nutrients and has a special role in maintaining redox homeostasis in physiological and physio-pathological brain states. Furthermore, because of its capacity to chelate metal ions, ascorbic acid may particularly serve as a potent therapeutic agent in Pb poisoning. The present review first discusses the major consequences of Pb exposure in children and then proceeds to present evidence from human and animal studies for ascorbic acid as an efficient ameliorative supplemental nutrient in Pb poisoning, with a particular focus on developmental Pb neurotoxicity. In doing so, it is hoped that there is a revitalization for further research on understanding the brain functions of this essential, safe, and readily available vitamin in physiological states, as well to justify and establish it as an effective neuroprotective and modulatory factor in the pathologies of the nervous system, including developmental neuropathologies.
Collapse
|
12
|
Lee KH, Cha M, Lee BH. Neuroprotective Effect of Antioxidants in the Brain. Int J Mol Sci 2020; 21:ijms21197152. [PMID: 32998277 PMCID: PMC7582347 DOI: 10.3390/ijms21197152] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/18/2020] [Accepted: 09/23/2020] [Indexed: 12/29/2022] Open
Abstract
The brain is vulnerable to excessive oxidative insults because of its abundant lipid content, high energy requirements, and weak antioxidant capacity. Reactive oxygen species (ROS) increase susceptibility to neuronal damage and functional deficits, via oxidative changes in the brain in neurodegenerative diseases. Overabundance and abnormal levels of ROS and/or overload of metals are regulated by cellular defense mechanisms, intracellular signaling, and physiological functions of antioxidants in the brain. Single and/or complex antioxidant compounds targeting oxidative stress, redox metals, and neuronal cell death have been evaluated in multiple preclinical and clinical trials as a complementary therapeutic strategy for combating oxidative stress associated with neurodegenerative diseases. Herein, we present a general analysis and overview of various antioxidants and suggest potential courses of antioxidant treatments for the neuroprotection of the brain from oxidative injury. This review focuses on enzymatic and non-enzymatic antioxidant mechanisms in the brain and examines the relative advantages and methodological concerns when assessing antioxidant compounds for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Kyung Hee Lee
- Department of Dental Hygiene, Division of Health Science, Dongseo University, Busan 47011, Korea;
| | - Myeounghoon Cha
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea;
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-2-2228-1711
| |
Collapse
|
13
|
Liu B, Li J, Lin X, Hu J, Lou S. The metabolic changes in the hippocampus of an atherosclerotic rat model and the regulation of aerobic training. Metab Brain Dis 2020; 35:1017-1034. [PMID: 32240489 DOI: 10.1007/s11011-020-00566-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 03/17/2020] [Indexed: 01/17/2023]
Abstract
Atherosclerosis has been associated with the progression of cognitive impairment and the effect of metabolic changes in the brain on cognitive function may be pronounced. The aim is to reveal the metabolic changes during atherosclerosis and clarify the possible role of exercise in regulating hippocampal metabolism. Hence, A rat model of atherosclerosis was established by high-fat diet feeding in combination with vitamin D3 intraperitoneal injection, then 4 weeks of aerobic exercise was conducted. Metabolomics based on GC-MS was applied to detect small molecules metabolites and western blot was used to detect the concentration of enzymes involved in metabolic changes in rat hippocampus. Compared to the control group, metabolites including xylulose 5-phosphate, threonine, succinate, and nonanoic acid were markedly elevated, whereas methyl arachidonic acid and methyl stearate decreased in the AS group, accompanied by a raised concentration of aldose reductase and glucose 6-phosphate dehydrogenase as well as a declined concentration of acetyl-CoA carboxylase and fatty acid synthase. After 4 weeks' aerobic exercise, the levels of succinic acid, branched chain amino acids, nonanoic acid, desmosterol, and aldose reductase decreased, whereas methyl arachidonic acid, methyl stearate, and glyceraldehyde-3-phosphate elevated in the hippocampus of the TAS group in comparison with the AS group. These results suggest that atherosclerosis could cause a severe metabolic disturbance, and aerobic exercise plays an important role in regulating atherosclerosis-induced disorder of glucose metabolism in the hippocampus.
Collapse
Affiliation(s)
- Beibei Liu
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China
- Department of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Jingjing Li
- Post-doctoral station of clinical medicine, Tongji Hospital, medical school of Tongji University, Shanghai, 200092, China
| | - Xiaojing Lin
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China
| | - Jingyun Hu
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China
| | - Shujie Lou
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
14
|
Gordon DS, Rudinsky AJ, Guillaumin J, Parker VJ, Creighton KJ. Vitamin C in Health and Disease: A Companion Animal Focus. Top Companion Anim Med 2020; 39:100432. [PMID: 32482285 DOI: 10.1016/j.tcam.2020.100432] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/31/2022]
Abstract
Vitamin C is synthesized in the liver in most species, including dogs and cats, and is widely distributed through body tissues. Vitamin C has an important physiologic role in numerous metabolic functions including tissue growth and maintenance, amelioration of oxidative stress, and immune regulation. It is also a co-factor in the production of important substances such as catecholamines and vasopressin. Decreased vitamin C levels have been documented in a wide variety of diseases, and in critically ill human patients may be associated with increased severity of disease and decreased survival. Intravenous supplementation with vitamin C has been proposed as a potential life-saving treatment in conditions such as septic shock, and results of small some human trials are promising. Data in companion in animals is very limited, but the possible benefits and , seemingly low risk of adverse effects , and the low cost of this treatment make vitamin C therapy a promising area of future investigation in critically ill dogs and cats.
Collapse
Affiliation(s)
- Daniel S Gordon
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus OH, USA
| | - Adam J Rudinsky
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus OH, USA
| | - Julien Guillaumin
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus OH, USA
| | - Valerie J Parker
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus OH, USA
| | - Karina J Creighton
- Departments of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus OH, USA.
| |
Collapse
|
15
|
Jezierska-Wozniak K, Sinderewicz E, Czelejewska W, Wojtacha P, Barczewska M, Maksymowicz W. Influence of Bone Marrow-Derived Mesenchymal Stem Cell Therapy on Oxidative Stress Intensity in Minimally Conscious State Patients. J Clin Med 2020; 9:E683. [PMID: 32138308 PMCID: PMC7141306 DOI: 10.3390/jcm9030683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 02/28/2020] [Indexed: 12/11/2022] Open
Abstract
Neurological disorders, including minimally conscious state (MCS), may be associated with the presence of high concentrations of reactive oxygen species within the central nervous system. Regarding the documented role of mesenchymal stem cells (MSCs) in oxidative stress neutralization, the aim of this study is to evaluate the effect of bone marrow-derived MSC (BM-MSC) transplantation on selected markers of oxidative stress in MCS patients. Antioxidant capacity was measured in cerebrospinal fluid (CSF) and plasma collected from nine patients aged between 19 and 45 years, remaining in MCS for 3 to 14 months. Total antioxidant capacity, ascorbic acid and ascorbate concentrations, superoxide dismutase, catalase, and peroxidase activity were analyzed and the presence of tested antioxidants in the CSF and plasma was confirmed. Higher ascorbic acid (AA) content and catalase (CAT) activity were noted in CSF relative to plasma, whereas superoxide dismutase (SOD) activity and total antioxidant capacity were higher in plasma relative to CSF. Total antioxidant capacity measured in CSF was greater after BM-MSC transplantations. The content of ascorbates was lower and CAT activity was higher both in CSF and plasma after the administration of BM-MSC. The above results suggest that MSCs modulate oxidative stress intensity in MCS patients, mainly via ascorbates and CAT activity.
Collapse
Affiliation(s)
- Katarzyna Jezierska-Wozniak
- Department of Neurosurgery, Laboratory of Regenerative Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska 30 Str., 10-082 Olsztyn, Poland; (E.S.); (W.C.)
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska 30 Str., 10-082 Olsztyn, Poland; (M.B.); (W.M.)
| | - Emilia Sinderewicz
- Department of Neurosurgery, Laboratory of Regenerative Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska 30 Str., 10-082 Olsztyn, Poland; (E.S.); (W.C.)
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska 30 Str., 10-082 Olsztyn, Poland; (M.B.); (W.M.)
| | - Wioleta Czelejewska
- Department of Neurosurgery, Laboratory of Regenerative Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska 30 Str., 10-082 Olsztyn, Poland; (E.S.); (W.C.)
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska 30 Str., 10-082 Olsztyn, Poland; (M.B.); (W.M.)
| | - Pawel Wojtacha
- Department of Industrial and Food Microbiology, Faculty of Food Sciences, University of Warmia and Mazury in Olsztyn, Plac Cieszynski 1 Str., 10-726 Olsztyn, Poland;
| | - Monika Barczewska
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska 30 Str., 10-082 Olsztyn, Poland; (M.B.); (W.M.)
| | - Wojciech Maksymowicz
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska 30 Str., 10-082 Olsztyn, Poland; (M.B.); (W.M.)
| |
Collapse
|
16
|
Basal Sodium-Dependent Vitamin C Transporter 2 polarization in choroid plexus explant cells in normal or scorbutic conditions. Sci Rep 2019; 9:14422. [PMID: 31594969 PMCID: PMC6783570 DOI: 10.1038/s41598-019-50772-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/15/2019] [Indexed: 01/10/2023] Open
Abstract
Vitamin C is incorporated into the cerebrospinal fluid (CSF) through choroid plexus cells. While the transfer of vitamin C from the blood to the brain has been studied functionally, the vitamin C transporter, SVCT2, has not been detected in the basolateral membrane of choroid plexus cells. Furthermore, it is unknown how its expression is induced in the developing brain and modulated in scurvy conditions. We concluded that SVCT2 is intensely expressed in the second half of embryonic brain development and postnatal stages. In postnatal and adult brain, SVCT2 is highly expressed in all choroidal plexus epithelial cells, shown by colocalization with GLUT1 in the basolateral membranes and without MCT1 colocalization, which is expressed in the apical membrane. We confirmed that choroid plexus explant cells (in vitro) form a sealed epithelial structure, which polarized basolaterally, endogenous or overexpressed SVCT2. These results are reproduced in vivo by injecting hSVCT2wt-EYFP lentivirus into the CSF. Overexpressed SVCT2 incorporates AA (intraperitoneally injected) from the blood to the CSF. Finally, we observed in Guinea pig brain under scorbutic condition, that normal distribution of SVCT2 in choroid plexus may be regulated by peripheral concentrations of vitamin C. Additionally, we observed that SVCT2 polarization also depends on the metabolic stage of the choroid plexus cells.
Collapse
|
17
|
Ballaz SJ, Rebec GV. Neurobiology of vitamin C: Expanding the focus from antioxidant to endogenous neuromodulator. Pharmacol Res 2019; 146:104321. [PMID: 31229562 DOI: 10.1016/j.phrs.2019.104321] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 01/06/2023]
Abstract
Ascorbic acid (AA) is a water-soluble vitamin (C) found in all bodily organs. Most mammals synthesize it, humans are required to eat it, but all mammals need it for healthy functioning. AA reaches its highest concentration in the brain where both neurons and glia rely on tightly regulated uptake from blood via the glucose transport system and sodium-coupled active transport to accumulate and maintain AA at millimolar levels. As a prototype antioxidant, AA is not only neuroprotective, but also functions as a cofactor in redox-coupled reactions essential for the synthesis of neurotransmitters (e.g., dopamine and norepinephrine) and paracrine lipid mediators (e.g., epoxiecoisatrienoic acids) as well as the epigenetic regulation of DNA. Although redox capacity led to the promotion of AA in high doses as potential treatment for various neuropathological and psychiatric conditions, ample evidence has not supported this therapeutic strategy. Here, we focus on some long-neglected aspects of AA neurobiology, including its modulatory role in synaptic transmission as demonstrated by the long-established link between release of endogenous AA in brain extracellular fluid and the clearance of glutamate, an excitatory amino acid. Evidence that this link can be disrupted in animal models of Huntington´s disease is revealing opportunities for new research pathways and therapeutic applications (e.g., epilepsy and pain management). In fact, we suggest that improved understanding of the regulation of endogenous AA and its interaction with key brain neurotransmitter systems, rather than administration of AA in excess, should be the target of future brain-based therapies.
Collapse
Affiliation(s)
- Santiago J Ballaz
- School of Biological Sciences and Engineering, Yachay Tech University, Urcuqui, Ecuador.
| | - George V Rebec
- Program in Neuroscience, Department Psychological & Brain Sciences, Indiana University, Bloomington, USA.
| |
Collapse
|
18
|
Ferrada L, Salazar K, Nualart F. Metabolic control by dehydroascorbic acid: Questions and controversies in cancer cells. J Cell Physiol 2019; 234:19331-19338. [DOI: 10.1002/jcp.28637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Luciano Ferrada
- Departamento de Biología Celular, Laboratorio de Neurobiología y células madres Neuro‐CellTT, Centro de Microscopía Avanzada CMA BIOBIO, Facultad de Ciencias Biológicas Universidad de Concepción Concepción Chile
| | - Katterine Salazar
- Departamento de Biología Celular, Laboratorio de Neurobiología y células madres Neuro‐CellTT, Centro de Microscopía Avanzada CMA BIOBIO, Facultad de Ciencias Biológicas Universidad de Concepción Concepción Chile
| | - Francisco Nualart
- Departamento de Biología Celular, Laboratorio de Neurobiología y células madres Neuro‐CellTT, Centro de Microscopía Avanzada CMA BIOBIO, Facultad de Ciencias Biológicas Universidad de Concepción Concepción Chile
| |
Collapse
|
19
|
Salazar K, Martínez F, Pérez-Martín M, Cifuentes M, Trigueros L, Ferrada L, Espinoza F, Saldivia N, Bertinat R, Forman K, Oviedo MJ, López-Gambero AJ, Bonansco C, Bongarzone ER, Nualart F. SVCT2 Expression and Function in Reactive Astrocytes Is a Common Event in Different Brain Pathologies. Mol Neurobiol 2017; 55:5439-5452. [PMID: 28942474 DOI: 10.1007/s12035-017-0762-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/31/2017] [Indexed: 11/28/2022]
Abstract
Ascorbic acid (AA), the reduced form of vitamin C, acts as a neuroprotector by eliminating free radicals in the brain. Sodium/vitamin C co-transporter isoform 2 (SVCT2) mediates uptake of AA by neurons. It has been reported that SVCT2 mRNA is induced in astrocytes under ischemic damage, suggesting that its expression is enhanced in pathological conditions. However, it remains to be established if SVCT expression is altered in the presence of reactive astrogliosis generated by different brain pathologies. In the present work, we demonstrate that SVCT2 expression is increased in astrocytes present at sites of neuroinflammation induced by intracerebroventricular injection of a GFP-adenovirus or the microbial enzyme, neuraminidase. A similar result was observed at 5 and 10 days after damage in a model of traumatic injury and in the hippocampus and cerebral cortex in the in vivo kindling model of epilepsy. Furthermore, we defined that cortical astrocytes maintained in culture for long periods acquire markers of reactive gliosis and express SVCT2, in a similar way as previously observed in situ. Finally, by means of second harmonic generation and 2-photon fluorescence imaging, we analyzed brain necropsied material from patients with Alzheimer's disease (AD), which presented with an accumulation of amyloid plaques. Strikingly, although AD is characterized by focalized astrogliosis surrounding amyloid plaques, SVCT2 expression at the astroglial level was not detected. We conclude that SVCT2 is heterogeneously induced in reactive astrogliosis generated in different pathologies affecting the central nervous system (CNS).
Collapse
Affiliation(s)
- Katterine Salazar
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.,Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepcion, Chile
| | - Fernando Martínez
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.,Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepcion, Chile
| | - Margarita Pérez-Martín
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Manuel Cifuentes
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Laura Trigueros
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile.,Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Luciano Ferrada
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Francisca Espinoza
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Natalia Saldivia
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Romina Bertinat
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Katherine Forman
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - María José Oviedo
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Antonio J López-Gambero
- Department of Cell Biology, Genetics and Physiology, University of Malaga, IBIMA, BIONAND, Andalusian Center for Nanomedicine and Biotechnology and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Malaga, Spain
| | - Christian Bonansco
- Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Gran Bretaña Avenida 1111, 2360102, Valparaíso, Chile
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA.,Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA BIO BIO, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile. .,Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160-C, Concepcion, Chile.
| |
Collapse
|
20
|
Raymond YCF, Glenda CSL, Meng LK. Effects of High Doses of Vitamin C on Cancer Patients in Singapore: Nine Cases. Integr Cancer Ther 2015; 15:197-204. [PMID: 26679971 DOI: 10.1177/1534735415622010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Introduction Intravenous high-dose vitamin C therapy is widely used in naturopathic and integrative oncology; however, a study reviewing its effects has never been performed in Singapore. This article serves to document administration of supportive vitamin C therapy for cancer patients in Singapore. METHODS The clinical response of 9 cancer patients of differing stages to the regular administration of large doses (25-100 g/d) of intravenous vitamin C (IVC; ascorbic acid) is outlined. Tumor pathology and patient health were verified by doctors who do not practice vitamin C treatment. RESULTS Cases suggesting survival beyond prognosis, improvement in quality of life, safe coadministration with and improved tolerance of conventional therapy, and deterioration in clinical condition following withdrawal of vitamin C therapy are documented clinically. Some patients experience the Jarisch-Herxheimer reaction-the release of endotoxin from microorganism death resulting in pimples, fever, and body odor-for a few hours after the therapy, but these are resolved quickly with no lasting effects. CONCLUSION Randomized trials of IVC therapy are recommended because it has minimal side effects and has shown promising results.
Collapse
Affiliation(s)
| | | | - Lim Kah Meng
- Gene Oasis Research and Innovation, Singapore Tianjin University, Tianjin 300072, PR China
| |
Collapse
|
21
|
Old Things New View: Ascorbic Acid Protects the Brain in Neurodegenerative Disorders. Int J Mol Sci 2015; 16:28194-217. [PMID: 26633354 PMCID: PMC4691042 DOI: 10.3390/ijms161226095] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/23/2015] [Accepted: 10/14/2015] [Indexed: 12/13/2022] Open
Abstract
Ascorbic acid is a key antioxidant of the Central Nervous System (CNS). Under brain activity, ascorbic acid is released from glial reservoirs to the synaptic cleft, where it is taken up by neurons. In neurons, ascorbic acid scavenges reactive oxygen species (ROS) generated during synaptic activity and neuronal metabolism where it is then oxidized to dehydroascorbic acid and released into the extracellular space, where it can be recycled by astrocytes. Other intrinsic properties of ascorbic acid, beyond acting as an antioxidant, are important in its role as a key molecule of the CNS. Ascorbic acid can switch neuronal metabolism from glucose consumption to uptake and use of lactate as a metabolic substrate to sustain synaptic activity. Multiple evidence links oxidative stress with neurodegeneration, positioning redox imbalance and ROS as a cause of neurodegeneration. In this review, we focus on ascorbic acid homeostasis, its functions, how it is used by neurons and recycled to ensure antioxidant supply during synaptic activity and how this antioxidant is dysregulated in neurodegenerative disorders.
Collapse
|
22
|
Dehydroascorbic Acid Promotes Cell Death in Neurons Under Oxidative Stress: a Protective Role for Astrocytes. Mol Neurobiol 2015; 53:5847-5863. [PMID: 26497038 DOI: 10.1007/s12035-015-9497-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/16/2015] [Indexed: 12/31/2022]
Abstract
Ascorbic acid (AA), the reduced form of vitamin C, is incorporated into neurons via the sodium ascorbate co-transporter SVCT2. However, this transporter is not expressed in astrocytes, which take up the oxidized form of vitamin C, dehydroascorbic acid (DHA), via the facilitative hexose transporter GLUT1. Therefore, neuron and astrocyte interactions are thought to mediate vitamin C recycling in the nervous system. Although astrocytes are essential for the antioxidant defense of neurons under oxidative stress, a condition in which a large amount of ROS is generated that may favor the extracellular oxidation of AA and the subsequent neuronal uptake of DHA via GLUT3, potentially increasing oxidative stress in neurons. This study analyzed the effects of oxidative stress and DHA uptake on neuronal cell death in vitro. Different analyses revealed the presence of the DHA transporters GLUT1 and GLUT3 in Neuro2a and HN33.11 cells and in cortical neurons. Kinetic analyses confirmed that all cells analyzed in this study possess functional GLUTs that take up 2-deoxyglucose and DHA. Thus, DHA promotes the death of stressed neuronal cells, which is reversed by incubating the cells with cytochalasin B, an inhibitor of DHA uptake by GLUT1 and GLUT3. Additionally, the presence of glial cells (U87 and astrocytes), which promote DHA recycling, reverses the observed cell death of stressed neurons. Taken together, these results indicate that DHA promotes the death of stressed neurons and that astrocytes are essential for the antioxidative defense of neurons. Thus, the astrocyte-neuron interaction may function as an essential mechanism for vitamin C recycling, participating in the antioxidative defense of the brain.
Collapse
|
23
|
Lane DJR, Richardson DR. The active role of vitamin C in mammalian iron metabolism: much more than just enhanced iron absorption! Free Radic Biol Med 2014; 75:69-83. [PMID: 25048971 DOI: 10.1016/j.freeradbiomed.2014.07.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/04/2014] [Accepted: 07/08/2014] [Indexed: 01/18/2023]
Abstract
Ascorbate is a cofactor in numerous metabolic reactions. Humans cannot synthesize ascorbate owing to inactivation of the gene encoding the enzyme l-gulono-γ-lactone oxidase, which is essential for ascorbate synthesis. Accumulating evidence strongly suggests that in addition to the known ability of dietary ascorbate to enhance nonheme iron absorption in the gut, ascorbate within mammalian systems can regulate cellular iron uptake and metabolism. Ascorbate modulates iron metabolism by stimulating ferritin synthesis, inhibiting lysosomal ferritin degradation, and decreasing cellular iron efflux. Furthermore, ascorbate cycling across the plasma membrane is responsible for ascorbate-stimulated iron uptake from low-molecular-weight iron-citrate complexes, which are prominent in the plasma of individuals with iron-overload disorders. Importantly, this iron-uptake pathway is of particular relevance to astrocyte brain iron metabolism and tissue iron loading in disorders such as hereditary hemochromatosis and β-thalassemia. Recent evidence also indicates that ascorbate is a novel modulator of the classical transferrin-iron uptake pathway, which provides almost all iron for cellular demands and erythropoiesis under physiological conditions. Ascorbate acts to stimulate transferrin-dependent iron uptake by an intracellular reductive mechanism, strongly suggesting that it may act to stimulate iron mobilization from the endosome. The ability of ascorbate to regulate transferrin iron uptake could help explain the metabolic defect that contributes to ascorbate-deficiency-induced anemia.
Collapse
Affiliation(s)
- Darius J R Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
24
|
Salazar K, Cerda G, Martínez F, Sarmiento JM, González C, Rodríguez F, García-Robles M, Tapia JC, Cifuentes M, Nualart F. SVCT2 transporter expression is post-natally induced in cortical neurons and its function is regulated by its short isoform. J Neurochem 2014; 130:693-706. [DOI: 10.1111/jnc.12793] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 05/28/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Katterine Salazar
- Laboratorio de Neurobiología y Células Madres; Facultad de Ciencias Biológicas; Centro de Microscopía Avanzada CMA-BIOBIO; Universidad de Concepción; Concepción Chile
| | - Gustavo Cerda
- Laboratorio de Neurobiología y Células Madres; Facultad de Ciencias Biológicas; Centro de Microscopía Avanzada CMA-BIOBIO; Universidad de Concepción; Concepción Chile
| | - Fernando Martínez
- Laboratorio de Neurobiología y Células Madres; Facultad de Ciencias Biológicas; Centro de Microscopía Avanzada CMA-BIOBIO; Universidad de Concepción; Concepción Chile
| | - José M. Sarmiento
- Departamento de Fisiología; Facultad de Medicina; Universidad Austral de Chile; Valdivia Chile
| | - Carlos González
- Departamento de Fisiología; Facultad de Medicina; Universidad Austral de Chile; Valdivia Chile
| | - Federico Rodríguez
- Laboratorio de Neurobiología y Células Madres; Facultad de Ciencias Biológicas; Centro de Microscopía Avanzada CMA-BIOBIO; Universidad de Concepción; Concepción Chile
| | - María García-Robles
- Laboratorio de Neurobiología y Células Madres; Facultad de Ciencias Biológicas; Centro de Microscopía Avanzada CMA-BIOBIO; Universidad de Concepción; Concepción Chile
| | - Juan Carlos Tapia
- Department of Neuroscience; Columbia University; New York city New York USA
| | - Manuel Cifuentes
- Departamento de Biología Celular, Genética y Fisiología; Facultad de Ciencias; Centro de Investigaciones Biomédicas en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN); Instituto de Investigación Biomédica de Málaga (IBIMA); Campus Universitario de Teatinos s/n; Universidad de Málaga; Málaga España
| | - Francisco Nualart
- Laboratorio de Neurobiología y Células Madres; Facultad de Ciencias Biológicas; Centro de Microscopía Avanzada CMA-BIOBIO; Universidad de Concepción; Concepción Chile
| |
Collapse
|
25
|
Acuña AI, Esparza M, Kramm C, Beltrán FA, Parra AV, Cepeda C, Toro CA, Vidal RL, Hetz C, Concha II, Brauchi S, Levine MS, Castro MA. A failure in energy metabolism and antioxidant uptake precede symptoms of Huntington's disease in mice. Nat Commun 2014; 4:2917. [PMID: 24336051 PMCID: PMC3905737 DOI: 10.1038/ncomms3917] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 11/12/2013] [Indexed: 01/22/2023] Open
Abstract
Huntington's disease has been associated with a failure in energy metabolism and oxidative damage. Ascorbic acid is a powerful antioxidant highly concentrated in the brain where it acts as a messenger, modulating neuronal metabolism. Using an electrophysiological approach in R6/2 HD slices, we observe an abnormal ascorbic acid flux from astrocytes to neurons, which is responsible for alterations in neuronal metabolic substrate preferences. Here using striatal neurons derived from knock-in mice expressing mutant huntingtin (STHdhQ cells), we study ascorbic acid transport. When extracellular ascorbic acid concentration increases, as occurs during synaptic activity, ascorbic acid transporter 2 (SVCT2) translocates to the plasma membrane, ensuring optimal ascorbic acid uptake for neurons. In contrast, SVCT2 from cells that mimic HD symptoms (dubbed HD cells) fails to reach the plasma membrane under the same conditions. We reason that an early impairment of ascorbic acid uptake in HD neurons could lead to early metabolic failure promoting neuronal death.
Collapse
Affiliation(s)
- Aníbal I Acuña
- 1] Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [2] Centro de Investigación Sur-Austral en Enfermedades del Sistema Nervioso (CISNe), Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [3] Escuela de Graduados, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [4]
| | - Magdalena Esparza
- 1] Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [2] Centro de Investigación Sur-Austral en Enfermedades del Sistema Nervioso (CISNe), Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [3]
| | - Carlos Kramm
- 1] Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [2] Centro de Investigación Sur-Austral en Enfermedades del Sistema Nervioso (CISNe), Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [3]
| | - Felipe A Beltrán
- 1] Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [2] Centro de Investigación Sur-Austral en Enfermedades del Sistema Nervioso (CISNe), Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [3] Escuela de Graduados, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile
| | - Alejandra V Parra
- 1] Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [2] Centro de Investigación Sur-Austral en Enfermedades del Sistema Nervioso (CISNe), Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, Brain Research Institute, The David Geffen School of Medicine, 760 Westwood Plaza, University of California Los Angeles, Los Angeles, California 90095-1759, USA
| | - Carlos A Toro
- 1] Escuela de Graduados, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [2] Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile
| | - René L Vidal
- Instituto de Ciencias Biomédicas, Universidad de Chile, Avda, Independencia 1027, Santiago, Chile
| | - Claudio Hetz
- Neurounion Biomedical Foundation, Independencia 1027, Santiago, Chile
| | - Ilona I Concha
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile
| | - Sebastián Brauchi
- 1] Centro de Investigación Sur-Austral en Enfermedades del Sistema Nervioso (CISNe), Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [2] Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile
| | - Michael S Levine
- Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, Brain Research Institute, The David Geffen School of Medicine, 760 Westwood Plaza, University of California Los Angeles, Los Angeles, California 90095-1759, USA
| | - Maite A Castro
- 1] Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile [2] Centro de Investigación Sur-Austral en Enfermedades del Sistema Nervioso (CISNe), Universidad Austral de Chile, Campus Isla Teja s/n, Valdivia, 5090000 Chile
| |
Collapse
|
26
|
Güney G, Kutlu HM, Genç L. Preparation and characterization of ascorbic acid loaded solid lipid nanoparticles and investigation of their apoptotic effects. Colloids Surf B Biointerfaces 2014; 121:270-80. [PMID: 24985762 DOI: 10.1016/j.colsurfb.2014.05.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 04/30/2014] [Accepted: 05/05/2014] [Indexed: 12/11/2022]
Abstract
In this paper, ascorbic acid (Vitamin C, AA) known as an antioxidant was successfully incorporated in solid lipid nanoparticles (SLNs) by hot homogenization and efficient delivery of AA to cancer cells. The obtained SLN formulations were characterized by Nano Zetasizer ZS and HPLC with the particle size being less than 250nm. AA-SLNs exhibited sustained release and high entrapment efficiency. According to MTT test results, AA-SLNs showed high cytotoxic activity compared to the free AA against H-Ras 5RP7 cells without damaging NIH/3T3 control cells. These results were supported by the Annexin V-PI and caspase-3 assay. Furthermore, as compared to the AA, AA-SLNs exhibited more efficient cellular uptake, accumulated in the cytoplasm and induced apoptosis which was observed by confocal laser scanning microscopy (CLSM) and transmission electron microscopy (TEM). Thus, the results of this study suggest that SLNs can be a potential nanocarrier system for AA.
Collapse
Affiliation(s)
- Gamze Güney
- Faculty of Medicine, Department of Medical Biology, Sakarya University, Sakarya, Turkey.
| | - H Mehtap Kutlu
- Faculty of Science, Department of Biology, Anadolu University, Eskişehir, Turkey
| | - Lütfi Genç
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Anadolu University, Eskişehir, Turkey; Plant, Drug and Scientific Researches Center (AUBIBAM), Anadolu University, Eskişehir, Turkey
| |
Collapse
|
27
|
Fritz H, Flower G, Weeks L, Cooley K, Callachan M, McGowan J, Skidmore B, Kirchner L, Seely D. Intravenous Vitamin C and Cancer: A Systematic Review. Integr Cancer Ther 2014; 13:280-300. [PMID: 24867961 DOI: 10.1177/1534735414534463] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Intravenous vitamin C (IVC) is a contentious adjunctive cancer therapy, widely used in naturopathic and integrative oncology settings. We conducted a systematic review of human interventional and observational studies assessing IVC for use in cancer patients. METHODS We searched MEDLINE, EMBASE, The Cochrane Library, CINAHL, and AMED from inception to April 2013 for human studies examining the safety, effectiveness, or pharmacokinetics of IVC use in cancer patients. RESULTS Of 897 records, a total of 39 reports of 37 studies were included: 2 randomized controlled trials (RCTs), 15 uncontrolled trials, 6 observational studies, and 14 case reports. IVC dosing ranged from 1 g to more than 200 g ascorbic acid per infusion, typically administered 2 to 3 times weekly. IVC does not appear to increase toxicity or interfere with antitumor effects of gemcitabine/erlotinib therapy or paclitaxel and carboplatin. Based on 1 RCT and data from uncontrolled human trials, IVC may improve time to relapse and possibly enhance reductions in tumor mass and improve survival in combination with chemotherapy. IVC may improve quality of life, physical function, and toxicities associated with chemotherapy, including fatigue, nausea, insomnia, constipation, and depression. Case reports document several instances of tumor regression and long-term disease-free survival associated with use of IVC. CONCLUSION There is limited high-quality clinical evidence on the safety and effectiveness of IVC. The existing evidence is preliminary and cannot be considered conclusive but is suggestive of a good safety profile and potentially important antitumor activity; however, more rigorous evidence is needed to conclusively demonstrate these effects. IVC may improve the quality of life and symptom severity of patients with cancer, and several cases of cancer remission have been reported. Well-designed, controlled studies of IVC therapy are needed.
Collapse
Affiliation(s)
- Heidi Fritz
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | - Gillian Flower
- Ottawa Integrative Cancer Centre, Ottawa, Ontario, Canada
| | - Laura Weeks
- Ottawa Integrative Cancer Centre, Ottawa, Ontario, Canada
| | - Kieran Cooley
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada University of Toronto, Toronto, Ontario, Canada
| | | | - Jessie McGowan
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | - Becky Skidmore
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | | | - Dugald Seely
- Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada Ottawa Integrative Cancer Centre, Ottawa, Ontario, Canada Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
28
|
Fiorani M, Azzolini C, Guidarelli A, Cerioni L, Cantoni O. A novel biological role of dehydroascorbic acid: Inhibition of Na(+)-dependent transport of ascorbic acid. Pharmacol Res 2014; 84:12-7. [PMID: 24769194 DOI: 10.1016/j.phrs.2014.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/14/2014] [Accepted: 04/14/2014] [Indexed: 12/01/2022]
Abstract
A U937 cell clone, in which low micromolar concentrations of ascorbic acid (AA) and dehydroascorbic acid (DHA) are taken up at identical rates, was used to investigate possible interactions between transport systems mediating cellular uptake of the two forms of the vitamin. Results obtained with different experimental approaches showed that DHA potently and reversibly inhibits AA uptake through Na(+)-AA cotransporters. Hence, a progressive increase in extracellular DHA concentrations in the presence of a fixed amount of AA caused an initial decrease in the net amount of vitamin C accumulated, and eventually, at higher levels, it caused an accumulation of the vitamin solely based on DHA uptake through hexose transporters. DHA-dependent inhibition of AA uptake was also detected in various other cell types. Taken together, our results provide evidence of a novel biological effect mediated by concentrations of DHA compatible with those produced at inflammatory sites.
Collapse
Affiliation(s)
- Mara Fiorani
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino 61029, Italy
| | - Catia Azzolini
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino 61029, Italy
| | - Andrea Guidarelli
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino 61029, Italy
| | - Liana Cerioni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino 61029, Italy
| | - Orazio Cantoni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino 61029, Italy.
| |
Collapse
|
29
|
Cisternas P, Silva-Alvarez C, Martínez F, Fernandez E, Ferrada L, Oyarce K, Salazar K, Bolaños JP, Nualart F. The oxidized form of vitamin C, dehydroascorbic acid, regulates neuronal energy metabolism. J Neurochem 2014; 129:663-71. [PMID: 24460956 DOI: 10.1111/jnc.12663] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 01/06/2014] [Accepted: 01/09/2014] [Indexed: 11/30/2022]
Abstract
Vitamin C is an essential factor for neuronal function and survival, existing in two redox states, ascorbic acid (AA), and its oxidized form, dehydroascorbic acid (DHA). Here, we show uptake of both AA and DHA by primary cultures of rat brain cortical neurons. Moreover, we show that most intracellular AA was rapidly oxidized to DHA. Intracellular DHA induced a rapid and dramatic decrease in reduced glutathione that was immediately followed by a spontaneous recovery. This transient decrease in glutathione oxidation was preceded by an increase in the rate of glucose oxidation through the pentose phosphate pathway (PPP), and a concomitant decrease in glucose oxidation through glycolysis. DHA stimulated the activity of glucose-6-phosphate dehydrogenase, the rate-limiting enzyme of the PPP. Furthermore, we found that DHA stimulated the rate of lactate uptake by neurons in a time- and dose-dependent manner. Thus, DHA is a novel modulator of neuronal energy metabolism by facilitating the utilization of glucose through the PPP for antioxidant purposes.
Collapse
Affiliation(s)
- Pedro Cisternas
- Laboratorio de Neurobiología, Departamento de Biología Celular, Centro de Microscopía Avanzada CMA BIOBIO, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Nualart F, Mack L, García A, Cisternas P, Bongarzone ER, Heitzer M, Jara N, Martínez F, Ferrada L, Espinoza F, Baeza V, Salazar K. Vitamin C Transporters, Recycling and the Bystander Effect in the Nervous System: SVCT2 versus Gluts. ACTA ACUST UNITED AC 2014; 4:209. [PMID: 25110615 PMCID: PMC4126260 DOI: 10.4172/2157-7633.1000209] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vitamin C is an essential micronutrient in the human diet; its deficiency leads to a number of symptoms and ultimately death. After entry into cells within the central nervous system (CNS) through sodium vitamin C transporters (SVCTs) and facilitative glucose transporters (GLUTs), vitamin C functions as a neuromodulator, enzymatic cofactor, and reactive oxygen species (ROS) scavenger; it also stimulates differentiation. In this review, we will compare the molecular and structural aspects of vitamin C and glucose transporters and their expression in endothelial or choroid plexus cells, which form part of the blood-brain barrier and blood-cerebrospinal fluid (CSF) barrier, respectively. Additionally, we will describe SVCT and GLUT expression in different cells of the brain as well as SVCT2 distribution in tanycytes and astrocytes of the hypothalamic region. Finally, we will describe vitamin C recycling in the brain, which is mediated by a metabolic interaction between astrocytes and neurons, and the role of the "bystander effect" in the recycling mechanism of vitamin C in both normal and pathological conditions.
Collapse
Affiliation(s)
- Francisco Nualart
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Lauren Mack
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Andrea García
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Pedro Cisternas
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois Chicago, USA
| | - Marjet Heitzer
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Nery Jara
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Fernando Martínez
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Luciano Ferrada
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Francisca Espinoza
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Victor Baeza
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| | - Katterine Salazar
- Center for Advanced Microscopy CMA BIO-BIO, Neurobiology and Stem cell Laboratory, Concepcion University, Chile
| |
Collapse
|
31
|
The glutamate aspartate transporter (GLAST) mediates L-glutamate-stimulated ascorbate-release via swelling-activated anion channels in cultured neonatal rodent astrocytes. Cell Biochem Biophys 2013; 65:107-19. [PMID: 22886112 DOI: 10.1007/s12013-012-9404-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vitamin C (ascorbate) plays important neuroprotective and neuromodulatory roles in the mammalian brain. Astrocytes are crucially involved in brain ascorbate homeostasis and may assist in regenerating extracellular ascorbate from its oxidised forms. Ascorbate accumulated by astrocytes can be released rapidly by a process that is stimulated by the excitatory amino acid, L-glutamate. This process is thought to be neuroprotective against excitotoxicity. Although of potential clinical interest, the mechanism of this stimulated ascorbate-release remains unknown. Here, we report that primary cultures of mouse and rat astrocytes release ascorbate following initial uptake of dehydroascorbate and accumulation of intracellular ascorbate. Ascorbate-release was not due to cellular lysis, as assessed by cellular release of the cytosolic enzyme lactate dehydrogenase, and was stimulated by L-glutamate and L-aspartate, but not the non-excitatory amino acid L-glutamine. This stimulation was due to glutamate-induced cellular swelling, as it was both attenuated by hypertonic and emulated by hypotonic media. Glutamate-stimulated ascorbate-release was also sensitive to inhibitors of volume-sensitive anion channels, suggesting that the latter may provide the conduit for ascorbate efflux. Glutamate-stimulated ascorbate-release was not recapitulated by selective agonists of either ionotropic or group I metabotropic glutamate receptors, but was completely blocked by either of two compounds, TFB-TBOA and UCPH-101, which non-selectively and selectively inhibit the glial Na(+)-dependent excitatory amino acid transporter, GLAST, respectively. These results suggest that an impairment of astrocytic ascorbate-release may exacerbate neuronal dysfunction in neurodegenerative disorders and acute brain injury in which excitotoxicity and/or GLAST deregulation have been implicated.
Collapse
|
32
|
Rodríguez FS, Salazar KA, Jara NA, García-Robles MA, Pérez F, Ferrada LE, Martínez F, Nualart FJ. Retracted: Superoxide-dependent uptake of vitamin C in human glioma cells. J Neurochem 2013; 127:793-804. [DOI: 10.1111/jnc.12365] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/25/2013] [Accepted: 07/08/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Federico S. Rodríguez
- Laboratory of Neurobiology and Stem Cells; Center for Advanced Microscopy CMA BIOBIO; University of Concepcion; Concepcion Chile
| | - Katterine A. Salazar
- Laboratory of Neurobiology and Stem Cells; Center for Advanced Microscopy CMA BIOBIO; University of Concepcion; Concepcion Chile
| | - Nery A. Jara
- Laboratory of Neurobiology and Stem Cells; Center for Advanced Microscopy CMA BIOBIO; University of Concepcion; Concepcion Chile
| | | | | | - Luciano E. Ferrada
- Laboratory of Neurobiology and Stem Cells; Center for Advanced Microscopy CMA BIOBIO; University of Concepcion; Concepcion Chile
| | - Fernando Martínez
- Laboratory of Neurobiology and Stem Cells; Center for Advanced Microscopy CMA BIOBIO; University of Concepcion; Concepcion Chile
- Laboratory of Cellular Biology; University of Concepcion; Concepcion Chile
| | - Francisco J. Nualart
- Laboratory of Neurobiology and Stem Cells; Center for Advanced Microscopy CMA BIOBIO; University of Concepcion; Concepcion Chile
| |
Collapse
|
33
|
Pastor P, Cisternas P, Salazar K, Silva-Alvarez C, Oyarce K, Jara N, Espinoza F, Martínez AD, Nualart F. SVCT2 vitamin C transporter expression in progenitor cells of the postnatal neurogenic niche. Front Cell Neurosci 2013; 7:119. [PMID: 23964197 PMCID: PMC3741466 DOI: 10.3389/fncel.2013.00119] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 07/08/2013] [Indexed: 12/20/2022] Open
Abstract
Known as a critical antioxidant, recent studies suggest that vitamin C plays an important role in stem cell generation, proliferation and differentiation. Vitamin C also enhances neural differentiation during cerebral development, a function that has not been studied in brain precursor cells. We observed that the rat neurogenic niche is structurally organized at day 15 of postnatal development, and proliferation and neural differentiation increase at day 21. In the human brain, a similar subventricular niche was observed at 1-month of postnatal development. Using immunohistochemistry, sodium-vitamin C cotransporter 2 (SVCT2) expression was detected in the subventricular zone (SVZ) and rostral migratory stream (RMS). Low co-distribution of SVCT2 and βIII-tubulin in neuroblasts or type-A cells was detected, and minimal co-localization of SVCT2 and GFAP in type-B or precursor cells was observed. Similar results were obtained in the human neurogenic niche. However, BrdU-positive cells also expressed SVCT2, suggesting a role of vitamin C in neural progenitor proliferation. Primary neurospheres prepared from rat brain and the P19 teratocarcinoma cell line, which forms neurospheres in vitro, were used to analyze the effect of vitamin C in neural stem cells. Both cell types expressed functional SVCT2 in vitro, and ascorbic acid (AA) induced their neural differentiation, increased βIII-tubulin and SVCT2 expression, and amplified vitamin C uptake.
Collapse
Affiliation(s)
- Patricia Pastor
- Laboratory of Neurobiology and Stem Cells, Department of Cellular Biology, Center for Advanced Microscopy CMA BIOBIO, University of Concepción Concepción, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bürzle M, Suzuki Y, Ackermann D, Miyazaki H, Maeda N, Clémençon B, Burrier R, Hediger MA. The sodium-dependent ascorbic acid transporter family SLC23. Mol Aspects Med 2013; 34:436-54. [PMID: 23506882 DOI: 10.1016/j.mam.2012.12.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/16/2012] [Indexed: 12/31/2022]
Affiliation(s)
- Marc Bürzle
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Nualart F, Castro T, Low M, Henríquez JP, Oyarce K, Cisternas P, García A, Yáñez AJ, Bertinat R, Montecinos VP, García-Robles MA. Dynamic expression of the sodium-vitamin C co-transporters, SVCT1 and SVCT2, during perinatal kidney development. Histochem Cell Biol 2012; 139:233-47. [PMID: 22990596 DOI: 10.1007/s00418-012-1027-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2012] [Indexed: 11/29/2022]
Abstract
Isoform 1 of the sodium-vitamin C co-transporter (SVCT1) is expressed in the apical membrane of proximal tubule epithelial cells in adult human and mouse kidneys. This study is aimed at analyzing the expression and function of SVCTs during kidney development. RT-PCR and immunohistochemical analyses revealed that SVCT1 expression is increased progressively during postnatal kidney development. However, SVCT1 transcripts were barely detected, if not absent, in the embryonic kidney. Instead, the high-affinity transporter, isoform 2 (SVCT2), was strongly expressed in the developing kidney from E15; its expression decreased at postnatal stages. Immunohistochemical analyses showed a dynamic distribution of SVCT2 in epithelial cells during kidney development. In renal cortex tubular epithelial cells, intracellular distribution of SVCT2 was observed at E19 with distribution in the basolateral membrane at P1. In contrast, SVCT2 was localized to the apical and basolateral membranes between E17 and E19 in medullary kidney tubular cells but was distributed intracellularly at P1. In agreement with these findings, functional expression of SVCT2, but not SVCT1 was detected in human embryonic kidney-derived (HEK293) cells. In addition, kinetic analysis suggested that an ascorbate-dependent mechanism accounts for targeted SVCT2 expression in the developing kidney during medullary epithelial cell differentiation. However, during cortical tubular differentiation, SVCT1 was induced and localized to the apical membrane of tubular epithelial cells. SVCT2 showed a basolateral polarization only for the first days of postnatal life. These studies suggest that the uptake of vitamin C mediated by different SVCTs plays differential roles during the ontogeny of kidney tubular epithelial cells.
Collapse
Affiliation(s)
- Francisco Nualart
- Departamento de Biología Celular, Centro de Microscopía Avanzada CMA BIO-BIO, Universidad de Concepción, Concepción, Chile.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bürzle M, Hediger MA. Functional and Physiological Role of Vitamin C Transporters. CO-TRANSPORT SYSTEMS 2012. [DOI: 10.1016/b978-0-12-394316-3.00011-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
37
|
|
38
|
Studies with low micromolar levels of ascorbic and dehydroascorbic acid fail to unravel a preferential route for vitamin C uptake and accumulation in U937 cells. Br J Nutr 2011; 107:691-6. [PMID: 21794197 DOI: 10.1017/s0007114511003540] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mammalian cells accumulate vitamin C either as ascorbic acid (AA), via Na+-AA co-transport, or dehydroascorbic acid (DHA, the oxidation product of AA), via facilitative hexose transport. As the latter, unlike the former, is a high-capacity transport mechanism, cultured cells normally accumulate greater levels of vitamin C when exposed to increasing concentrations of DHA as compared with AA. We report herein similar results using the U937 cell clone used in our laboratory only under conditions in which DHA and AA are used at concentrations greater than 50-60 μm. Below 60 μm, i.e. at levels in which AA is normally found in most biological fluids, AA and DHA are in fact taken up with identical rates and kinetics. Consequently, extracellular oxidation of AA switches the mode of uptake with hardly any effect on the net amount of vitamin C accumulated. As a final note, under these conditions, neither AA nor DHA causes detectable toxicity or any change in the redox status of the cells, as assessed by the reduced glutathione/reduced pyridine nucleotide pool. These findings therefore imply that some cell types do not have a preferential route for vitamin C accumulation, and that the uptake mechanism is uniquely dependent on the extracellular availability of AA v. DHA.
Collapse
|
39
|
Gomes ME, El Messaoudi S, Lenders JWM, Bellersen L, Verheugt FWA, Smits P, Tack CJ. High dose ascorbic acid does not reverse central sympathetic overactivity in chronic heart failure. J Clin Pharm Ther 2010; 36:546-52. [PMID: 21916906 DOI: 10.1111/j.1365-2710.2010.01205.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE The increased central sympathetic activity typically associated with chronic heart failure (CHF) is probably mediated by formation of reactive oxygen species (ROS) in the brain. Our objective was to undertake a trial to test our hypothesis that administration of the well-known antioxidant and ROS scavenger ascorbic acid, would reverse or reduce the sympathetic overactivity in CHF patients. METHODS In a prospective, randomized, placebo-controlled, double-blind, cross-over trial, 11 CHF patients were treated with ascorbic acid 2 g/day or placebo for 3 days. At the end of each treatment period, sympathetic nervous system activity was measured by microneurography for direct muscle sympathetic nerve activity (MSNA) recording, analysis of heart rate variability (HRV) and measurement of plasma norepinephrine concentrations. RESULTS During ascorbic acid administration, plasma vitamin C levels were higher than during placebo (74·9 ± 6·0 μmol/L vs. 54·8 ± 4·6 μmol/L, P = 0·03). Ascorbic acid had no effect on sympathetic activity: MSNA (ascorbic acid: 66·8 ± 3·3 vs. placebo 66·9 ± 3·2 bursts/100 beats, P = 0·98). In addition, HRV and plasma norepinephrine levels did not differ. WHAT IS NEW AND CONCLUSION Short-term administration of the antioxidant ascorbic acid in CHF patients does not reverse the increased sympathetic activity as measured by microneurography, HRV and plasma norepinephrine levels. The use of higher oral dosages seems not feasible due to accompanying side effects.
Collapse
Affiliation(s)
- M E Gomes
- Department of Internal Medicine Department of Cardiology Department of Pharmacology Toxicology, University Medical Center Nijmegen, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
40
|
Tveden-Nyborg P, Johansen LK, Raida Z, Villumsen CK, Larsen JO, Lykkesfeldt J. Vitamin C deficiency in early postnatal life impairs spatial memory and reduces the number of hippocampal neurons in guinea pigs. Am J Clin Nutr 2009; 90:540-6. [PMID: 19640959 DOI: 10.3945/ajcn.2009.27954] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The neonatal brain is particularly vulnerable to imbalances in redox homeostasis because of rapid growth and immature antioxidant systems. Vitamin C has been shown to have a key function in the brain, and during states of deficiency it is able to retain higher concentrations of vitamin C than other organs. However, because neurons maintain one of the highest intracellular concentrations of vitamin C in the organism, the brain may still be more sensitive to deficiency despite these preventive measures. OBJECTIVE The objective was to study the potential link between chronic vitamin C deficiency and neuronal damage in newborn guinea pigs. DESIGN Thirty 6- to 7-d-old guinea pigs were randomly assigned to 2 groups to receive either a vitamin C-sufficient diet or the same diet containing a low concentration of vitamin C (but adequate to prevent scurvy) for 2 mo. Spatial memory was assessed by the Morris Water Maze, and hippocampal neuron numbers were quantified by stereologic techniques. RESULTS The results showed a reduction in spatial memory (P < 0.05) and an increased time to first platform hit (P < 0.05) in deficient animals compared with controls. The deficient animals had a lower total number of neurons in hippocampal subdivisions (dentate gyrus, cornu ammonis 1, and cornu ammonis 2-3) than did the normal controls (P < 0.05). CONCLUSIONS Our data show that vitamin C deficiency in early postnatal life results in impaired neuronal development and a functional decrease in spatial memory in guinea pigs. We speculate that this unrecognized effect of vitamin C deficiency may have clinical implications for high-risk individuals, such as in children born from vitamin C-deficient mothers.
Collapse
|
41
|
Castro MA, Beltrán FA, Brauchi S, Concha II. A metabolic switch in brain: glucose and lactate metabolism modulation by ascorbic acid. J Neurochem 2009; 110:423-40. [PMID: 19457103 DOI: 10.1111/j.1471-4159.2009.06151.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In this review, we discuss a novel function of ascorbic acid in brain energetics. It has been proposed that during glutamatergic synaptic activity neurons preferably consume lactate released from glia. The key to this energetic coupling is the metabolic activation that occurs in astrocytes by glutamate and an increase in extracellular [K(+)]. Neurons are cells well equipped to consume glucose because they express glucose transporters and glycolytic and tricarboxylic acid cycle enzymes. Moreover, neuronal cells express monocarboxylate transporters and lactate dehydrogenase isoenzyme 1, which is inhibited by pyruvate. As glycolysis produces an increase in pyruvate concentration and a decrease in NAD(+)/NADH, lactate and glucose consumption are not viable at the same time. In this context, we discuss ascorbic acid participation as a metabolic switch modulating neuronal metabolism between rest and activation periods. Ascorbic acid is highly concentrated in CNS. Glutamate stimulates ascorbic acid release from astrocytes. Ascorbic acid entry into neurons and within the cell can inhibit glucose consumption and stimulate lactate transport. For this switch to occur, an ascorbic acid flow is necessary between astrocytes and neurons, which is driven by neural activity and is part of vitamin C recycling. Here, we review the role of glucose and lactate as metabolic substrates and the modulation of neuronal metabolism by ascorbic acid.
Collapse
Affiliation(s)
- Maite A Castro
- Instituto de Bioquímica, Universidad Austral de Chile, Valdivia, Chile.
| | | | | | | |
Collapse
|
42
|
Harrison FE, May JM. Vitamin C function in the brain: vital role of the ascorbate transporter SVCT2. Free Radic Biol Med 2009; 46:719-30. [PMID: 19162177 PMCID: PMC2649700 DOI: 10.1016/j.freeradbiomed.2008.12.018] [Citation(s) in RCA: 406] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 12/29/2008] [Accepted: 12/30/2008] [Indexed: 02/07/2023]
Abstract
Ascorbate (vitamin C) is a vital antioxidant molecule in the brain. However, it also has a number of other important functions, participating as a cofactor in several enzyme reactions, including catecholamine synthesis, collagen production, and regulation of HIF-1 alpha. Ascorbate is transported into the brain and neurons via the sodium-dependent vitamin C transporter 2 (SVCT2), which causes accumulation of ascorbate within cells against a concentration gradient. Dehydroascorbic acid, the oxidized form of ascorbate, is transported via glucose transporters of the GLUT family. Once in cells, it is rapidly reduced to ascorbate. The highest concentrations of ascorbate in the body are found in the brain and in neuroendocrine tissues such as adrenal, although the brain is the most difficult organ to deplete of ascorbate. Combined with regional asymmetry in ascorbate distribution within different brain areas, these facts suggest an important role for ascorbate in the brain. Ascorbate is proposed as a neuromodulator of glutamatergic, dopaminergic, cholinergic, and GABAergic transmission and related behaviors. Neurodegenerative diseases typically involve high levels of oxidative stress and thus ascorbate has been posited to have potential therapeutic roles against ischemic stroke, Alzheimer's disease, Parkinson's disease, and Huntington's disease.
Collapse
Affiliation(s)
| | - James M. May
- To whom correspondence should be addressed: Dr. James May, 7465 Medical Research Building IV, Vanderbilt University School of Medicine, Nashville, TN 37232-0475. Tel. (615) 936-1653; Fax: (615) 936-1667. E-mail:
| |
Collapse
|
43
|
Caprile T, Salazar K, Astuya A, Cisternas P, Silva-Alvarez C, Montecinos H, Millán C, García MDLA, Nualart F. The Na+-dependent l-ascorbic acid transporter SVCT2 expressed in brainstem cells, neurons, and neuroblastoma cells is inhibited by flavonoids. J Neurochem 2009; 108:563-77. [DOI: 10.1111/j.1471-4159.2008.05788.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
44
|
Portugal CC, Miya VS, Calaza KDC, Santos RAM, Paes-de-Carvalho R. Glutamate receptors modulate sodium-dependent and calcium-independent vitamin C bidirectional transport in cultured avian retinal cells. J Neurochem 2009; 108:507-20. [DOI: 10.1111/j.1471-4159.2008.05786.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
45
|
Differential distribution of the Sodium-vitamin C cotransporter-1 along the proximal tubule of the mouse and human kidney. Kidney Int 2008; 74:1278-86. [DOI: 10.1038/ki.2008.329] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Hosoya KI, Nakamura G, Akanuma SI, Tomi M, Tachikawa M. Dehydroascorbic acid uptake and intracellular ascorbic acid accumulation in cultured Müller glial cells (TR-MUL). Neurochem Int 2008; 52:1351-7. [DOI: 10.1016/j.neuint.2008.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 01/31/2008] [Accepted: 02/07/2008] [Indexed: 10/22/2022]
|
47
|
Ascorbic acid participates in a general mechanism for concerted glucose transport inhibition and lactate transport stimulation. Pflugers Arch 2008; 457:519-28. [PMID: 18506475 DOI: 10.1007/s00424-008-0526-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 04/24/2008] [Indexed: 12/31/2022]
Abstract
In this paper, we present a novel function for ascorbic acid. Ascorbic acid is an important water-soluble antioxidant and cofactor in various enzyme systems. We have previously demonstrated that an increase in neuronal intracellular ascorbic acid is able to inhibit glucose transport in cortical and hippocampal neurons. Because of the presence of sodium-dependent vitamin C transporters, ascorbic acid is highly concentrated in brain, testis, lung, and adrenal glands. In this work, we explored how ascorbic acid affects glucose and lactate uptake in neuronal and non-neuronal cells. Using immunofluorescence and reverse transcriptase-polymerase chain reaction (RT-PCR) analysis, the expression of glucose and ascorbic acid transporters in non-neuronal cells was studied. Like neurons, HEK293 cells expressed GLUT1, GLUT3, and SVCT2. With radioisotope-based methods, only intracellular ascorbic acid, but not extracellular, inhibits 2-deoxyglucose transport in HEK293 cells. As monocarboxylates such as pyruvate and lactate, are important metabolic sources, we analyzed the ascorbic acid effect on lactate transport in cultured neurons and HEK293 cells. Intracellular ascorbic acid was able to stimulate lactate transport in both cell types. Extracellular ascorbic acid did not affect this transport. Our data show that ascorbic acid inhibits glucose transport and stimulates lactate transport in neuronal and non-neuronal cells. Mammalian cells frequently present functional glucose and monocarboxylate transporters, and we describe here a general effect in which ascorbic acid functions like a glucose/monocarboxylate uptake switch in tissues expressing ascorbic acid transporters.
Collapse
|
48
|
Qiao H, May JM. Development of ascorbate transporters in brain cortical capillary endothelial cells in culture. Brain Res 2008; 1208:79-86. [PMID: 18394593 DOI: 10.1016/j.brainres.2008.02.102] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 02/27/2008] [Accepted: 02/27/2008] [Indexed: 11/24/2022]
Abstract
Ascorbic acid in its reduced form is not transported across the capillary endothelial cell blood-brain barrier. This is thought to be due to absence of the SVCT2, a specific transporter for ascorbate. To assess this directly we prepared primary cultures of mouse cortical microvascular endothelial cells. When still in the capillaries, these cells did not express the SVCT2 protein as assessed by immunocytochemistry and by immunoblotting. However, during several days in culture, they developed SVCT2 expression and showed ascorbate transport rates comparable to those in immortalized endothelial cell lines. SVCT2 expression was inversely proportional to cell density, was enhanced by culture at low physiologic plasma ascorbate concentrations, was inhibited by ascorbate concentrations expected in the brain interstitium, and was stimulated by cobalt ions. Expression of the SVCT2 was associated with ascorbate-dependent maturation and release of type IV collagen by the cells in culture. Although the SVCT2 is induced by culture of cortical capillary endothelial cells, its absence in vivo remains perplexing, given the need for intracellular ascorbate to facilitate type IV collagen maturation and release by endothelial cells.
Collapse
Affiliation(s)
- Huan Qiao
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-0475, USA
| | | |
Collapse
|
49
|
Luo S, Wang Z, Kansara V, Pal D, Mitra AK. Activity of a sodium-dependent vitamin C transporter (SVCT) in MDCK-MDR1 cells and mechanism of ascorbate uptake. Int J Pharm 2008; 358:168-76. [PMID: 18417304 DOI: 10.1016/j.ijpharm.2008.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 02/27/2008] [Accepted: 03/03/2008] [Indexed: 10/22/2022]
Abstract
The objective of this research was to functionally characterize sodium-dependent vitamin C transporter (SVCT) in MDCK-MDR1 cells and to study the effect of substituted benzene derivatives on the intracellular accumulation of ascorbic acid (AA). Mechanism of AA uptake and transport was delineated. Uptake of [(14)C]ascorbic acid ([(14)C]AA) was studied in the absence and presence of excess unlabelled AA, anion transporter inhibitors, and a series of mono- and di-substituted benzenes. Transepithelial transport of [(14)C]AA across polarized cell membrane has been studied for the first time. Role of cellular protein kinase-mediated pathways on the regulation of AA uptake has been investigated. The cellular localizations of SVCTs were observed using confocal microscopy. Uptake of AA was found to be saturable with a K(m) of 83.2muM and V(max) of 94.2pmol/min/mg protein for SVCT1. The process was pH, sodium, temperature, and energy-dependent. It was under the regulation of cellular protein kinase C (PKC) and Ca(2+)/CaM mediated pathways. [(14)C]AA uptake was significantly inhibited in the presence of excess unlabelled AA and a series of electron-withdrawing group, i.e., halogen- and nitro-substituted benzene derivatives. AA appears to translocate across polarized cell membrane from apical to basal side (A-B) as well as basal to apical side (B-A) at a similar permeability. It appears that SVCT1 was mainly expressed on the apical side and SVCT2 may be located on both apical and basal sides. In conclusion, SVCT has been functionally characterized in MDCK-MDR1 cells. The interference of a series of electrophile-substituted benzenes on the AA uptake process may be explained by their structural similarity. SVCT may be targeted to facilitate the delivery of drugs with low bioavailability by conjugating with AA and its structural analogs. MDCK-MDR1 cell line may be utilized as an in vitro model to study the permeability of AA conjugated prodrugs.
Collapse
Affiliation(s)
- Shuanghui Luo
- Division of Pharmaceutical Science, School of Pharmacy, University of Missouri-Kansas City, 5005 Rockhill Road, Kansas City, MO 64110-2499, USA
| | | | | | | | | |
Collapse
|
50
|
Castro MA, Pozo M, Cortés C, García MDLA, Concha II, Nualart F. Intracellular ascorbic acid inhibits transport of glucose by neurons, but not by astrocytes. J Neurochem 2007; 102:773-82. [PMID: 17630983 DOI: 10.1111/j.1471-4159.2007.04631.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It has been demonstrated that glutamatergic activity induces ascorbic acid (AA) depletion in astrocytes. Additionally, different data indicate that AA may inhibit glucose accumulation in primary cultures of rat hippocampal neurons. Thus, our hypothesis postulates that AA released from the astrocytes during glutamatergic synaptic activity may inhibit glucose uptake by neurons. We observed that cultured neurons express the sodium-vitamin C cotransporter 2 and the facilitative glucose transporters (GLUT) 1 and 3, however, in hippocampal brain slices GLUT3 was the main transporter detected. Functional activity of GLUTs was confirmed by means of kinetic analysis using 2-deoxy-d-glucose. Therefore, we showed that AA, once accumulated inside the cell, inhibits glucose transport in both cortical and hippocampal neurons in culture. Additionally, we showed that astrocytes are not affected by AA. Using hippocampal slices, we observed that upon blockade of monocarboxylate utilization by alpha-cyano-4-hydroxycinnamate and after glucose deprivation, glucose could rescue neuronal response to electrical stimulation only if AA uptake is prevented. Finally, using a transwell system of separated neuronal and astrocytic cultures, we observed that glutamate can reduce glucose transport in neurons only in presence of AA-loaded astrocytes, suggesting the essential role of astrocyte-released AA in this effect.
Collapse
Affiliation(s)
- Maite A Castro
- Instituto de Bioquímica, Facultad de Ciencias Biológicas, Universidad Austral de Chile, Valdivia, Chile
| | | | | | | | | | | |
Collapse
|