1
|
Fatima S, Alrashoudi RH, Alqarni SS, Alshehri S, Alsaigh SM, Malik A, Siddiqi NJ, Umrani A. Vitamin C Ameliorates Potassium Dichromate-Induced Oxidative Stress and Mitochondrial Dysfunction via PGC-1α/Nrf-2/TFAM Pathway. J Biochem Mol Toxicol 2025; 39:e70061. [PMID: 39714218 DOI: 10.1002/jbt.70061] [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: 04/13/2024] [Revised: 10/19/2024] [Accepted: 11/05/2024] [Indexed: 12/24/2024]
Abstract
Exposure to potassium dichromate (K2Cr2O7) is well known for its nephrotoxic effects on humans and animals. This study investigated the protective effects of vitamin C against K2Cr2O7-induced nephrotoxicity, focusing on its impact on altered carbohydrate metabolism, mitochondrial dysfunction, and associated molecular mechanisms in the cortical and medullary kidney segments. Male Wistar rats (n = 8) were divided into four groups: Group I received saline, Group II received a single 250 mg/kg body weight (bwt) intraperitoneal (i.p.) injection of vitamin C, Group III received K2Cr2O7 (15 mg/kg bwt, i.p.), and Group IV received vitamin C 6 h before K2Cr2O7 administration. Vitamin C significantly mitigated K2Cr2O7-induced nephrotoxic effects, restoring normal renal function and histological architecture. It preserved the activities of glycolytic and gluconeogenic enzymes altered by K2Cr2O7. Additionally, vitamin C mitigated K2Cr2O7-induced mitochondrial dysfunction by maintaining tricarboxylic acid (TCA) cycle enzymes, electron transport chain proteins, mitochondrial DNA copy number, and ATP content. It also reduced oxidative stress markers and enhanced antioxidant enzyme activity. The protective mechanism of vitamin C against K2Cr2O7-induced renal damage involved upregulation of the protein expression of peroxisome proliferation-activated receptor-γ coactivator-1α (PGC-1α), which further elevated the protein expression of nuclear factor erythroid 2-related factor-2 (Nrf-2) and transcription factor A, mitochondrial (TFAM), crucial for protecting cells from oxidative stress, enhancing mitochondrial function, and promoting cellular health. Overall, this study highlights the significant protective role of vitamin C against K2Cr2O7-induced renal damage by preserving carbohydrate metabolism and mitigating mitochondrial dysfunction through the PGC-1α/Nrf-2/TFAM pathway, offering valuable insights into its protective mechanisms in nephrotoxicity.
Collapse
Affiliation(s)
- Sabiha Fatima
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Reem H Alrashoudi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Sana S Alqarni
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Samiyah Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sara M Alsaigh
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Abdul Malik
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nikhat J Siddiqi
- Department of Internal Surgical Nursing, College of Nursing, King Saud University, Riyadh, Saudi Arabia
| | - Arbila Umrani
- Department of Anatomy, College of Medicine, King Khalid University Hospital, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Vaishampayan P, Lee Y. Redox-active vitamin C suppresses human osteosarcoma growth by triggering intracellular ROS-iron-calcium signaling crosstalk and mitochondrial dysfunction. Redox Biol 2024; 75:103288. [PMID: 39083898 PMCID: PMC11342202 DOI: 10.1016/j.redox.2024.103288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
Pharmacological vitamin C (VC) has gained attention for its pro-oxidant characteristics and selective ability to induce cancer cell death. However, defining its role in cancer has been challenging due to its complex redox properties. In this study, using a human osteosarcoma (OS) model, we show that the redox-active property of VC is critical for inducing non-apoptotic cancer cell death via intracellular reactive oxygen species (ROS)-iron-calcium crosstalk and mitochondrial dysfunction. In both 2D and 3D OS cell culture models, only the oxidizable form of VC demonstrated potent dose-dependent cytotoxicity, while non-oxidizable and oxidized VC derivatives had minimal effects. Live-cell imaging showed that only oxidizable VC caused a surge in cytotoxic ROS, dependent on iron rather than copper. Inhibitors of ferroptosis, a form of iron-dependent cell death, along with classical apoptosis inhibitors, were unable to completely counteract the cytotoxic effects induced by VC. Further pharmacological and genetic inhibition analyses showed that VC triggers calcium release through inositol 1,4,5-trisphosphate receptors (IP3Rs), leading to mitochondrial ROS production and eventual cell death. RNA sequencing revealed down-regulation of genes involved in the mitochondrial electron transport chain and oxidative phosphorylation upon pharmacological VC treatment. Consistently, high-dose VC reduced mitochondrial membrane potential, oxidative phosphorylation, and ATP levels, with ATP reconstitution rescuing VC-induced cytotoxicity. In vivo OS xenograft studies demonstrated reduced tumor growth with high-dose VC administration, concomitant with the altered expression of mitochondrial ATP synthase (MT-ATP). These findings emphasize VC's potential clinical utility in osteosarcoma treatment by inducing mitochondrial metabolic dysfunction through a vicious intracellular ROS-iron-calcium cycle.
Collapse
Affiliation(s)
- Prajakta Vaishampayan
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA
| | - Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA; Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, 99202, USA; Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
3
|
Shamel M, Baz S, Mahmoud H, Taghyan SA, Bakr MM, Al Ankily M. Balancing Risks versus Benefits: Vitamin C Therapy versus Copper Oxide Nanoparticles Toxicity in Albino Rats' Submandibular Salivary Gland. Eur J Dent 2024. [PMID: 38788769 DOI: 10.1055/s-0044-1786867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024] Open
Abstract
OBJECTIVES This study aimed to examine the suppressive effect of the natural antioxidant vitamin C (VC) against submandibular gland toxicity induced by copper oxide nanoparticles (CuO-NPs). MATERIALS AND METHODS Three groups of 30 mature male albino rats (4 weeks old) weighing between 150 and 200 g were selected. The rats were randomly assigned for 6 weeks to receive: intraperitoneal injection (IP) of vehicle (control group); IP of 2.5 mg/kg body weight (bw) of CuO-NPs (CuO-NPs group); and IP of 2.5 mg/kg bw of CuO-NPs, combined with a daily oral dose of 100 mg/kg bw of VC in drinking water via gavage (CuO-NPs/VC group). The rats were euthanized, and their submandibular glands were dissected for histological evaluation, including hematoxylin and eosin staining and immunohistochemistry for Ki-67 and caspase-3. STATISTICAL ANALYSIS The area expression for Ki-67 and caspase-3 was statistically analyzed using GraphPad Prism. Following analysis of variance analysis, Tukey's post hoc was used for multiple comparisons. The significance level was set at p < 0.05. RESULTS CuO-NPs caused significant cytotoxic effects on submandibular salivary gland cells in albino rats. This led to an increase in Ki-67 and caspase-3 levels compared with the control group. VC administration improved tissue histology and reduced Ki-67 and caspase-3 levels in the VC/CuO-NPs group compared with rats treated with CuO-NPs alone. CONCLUSION The study revealed significant cytotoxic effects of CuO-NPs on the submandibular salivary gland of albino rats. VC effectively mitigated these toxic effects, suggesting its potential as a readily available antioxidant.
Collapse
Affiliation(s)
- Mohamed Shamel
- Department of Oral Biology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Safaa Baz
- Department of Oral Pathology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Heba Mahmoud
- Department of Oral Biology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Salma Awad Taghyan
- Department of Oral Biology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| | - Mahmoud M Bakr
- General Dental Practice, School of Medicine and Dentistry, Griffith University, Gold Coast, Queensland, Australia
| | - Mahmoud Al Ankily
- Department of Oral Biology, Faculty of Dentistry, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
4
|
Portugal CC. Ascorbate and its transporter SVCT2: The dynamic duo's integrated roles in CNS neurobiology and pathophysiology. Free Radic Biol Med 2024; 212:448-462. [PMID: 38182073 DOI: 10.1016/j.freeradbiomed.2023.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/11/2023] [Accepted: 12/27/2023] [Indexed: 01/07/2024]
Abstract
Ascorbate is a small antioxidant molecule essential for the proper development and function of the brain. Ascorbate is transported into the brain and between brain cells via the Sodium vitamin C co-transporter 2 (SVCT2). This review provides an in-depth analysis of ascorbate's physiology, including how ascorbate is absorbed from food into the CNS, emphasizing cellular mechanisms of ascorbate recycling and release in different CNS compartments. Additionally, the review delves into the various functions of ascorbate in the CNS, including its impact on epigenetic modulation, synaptic plasticity, and neurotransmission. It also emphasizes ascorbate's role on neuromodulation and its involvement in neurodevelopmental processes and disorders. Furthermore, it analyzes the relationship between the duo ascorbate/SVCT2 in neuroinflammation, particularly its effects on microglial activation, cytokine release, and oxidative stress responses, highlighting its association with neurodegenerative diseases, such as Alzheimer's disease (AD). Overall, this review emphasizes the crucial role of the dynamic duo ascorbate/SVCT2 in CNS physiology and pathology and the need for further research to fully comprehend its significance in a neurobiological context and its potential therapeutic applications.
Collapse
Affiliation(s)
- Camila C Portugal
- I3s - Instituto de Investigação e Inovação em Saúde da Universidade do Porto and IBMC - Instituto de Biologia Molecular e Celular, Rua Alfredo Allen 208, 4200-135, Porto, Portugal.
| |
Collapse
|
5
|
Tempone MH, Borges-Martins VP, César F, Alexandrino-Mattos DP, de Figueiredo CS, Raony Í, dos Santos AA, Duarte-Silva AT, Dias MS, Freitas HR, de Araújo EG, Ribeiro-Resende VT, Cossenza M, P. Silva H, P. de Carvalho R, Ventura ALM, Calaza KC, Silveira MS, Kubrusly RCC, de Melo Reis RA. The Healthy and Diseased Retina Seen through Neuron-Glia Interactions. Int J Mol Sci 2024; 25:1120. [PMID: 38256192 PMCID: PMC10817105 DOI: 10.3390/ijms25021120] [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: 12/21/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
The retina is the sensory tissue responsible for the first stages of visual processing, with a conserved anatomy and functional architecture among vertebrates. To date, retinal eye diseases, such as diabetic retinopathy, age-related macular degeneration, retinitis pigmentosa, glaucoma, and others, affect nearly 170 million people worldwide, resulting in vision loss and blindness. To tackle retinal disorders, the developing retina has been explored as a versatile model to study intercellular signaling, as it presents a broad neurochemical repertoire that has been approached in the last decades in terms of signaling and diseases. Retina, dissociated and arranged as typical cultures, as mixed or neuron- and glia-enriched, and/or organized as neurospheres and/or as organoids, are valuable to understand both neuronal and glial compartments, which have contributed to revealing roles and mechanisms between transmitter systems as well as antioxidants, trophic factors, and extracellular matrix proteins. Overall, contributions in understanding neurogenesis, tissue development, differentiation, connectivity, plasticity, and cell death are widely described. A complete access to the genome of several vertebrates, as well as the recent transcriptome at the single cell level at different stages of development, also anticipates future advances in providing cues to target blinding diseases or retinal dysfunctions.
Collapse
Affiliation(s)
- Matheus H. Tempone
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Vladimir P. Borges-Martins
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Felipe César
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Dio Pablo Alexandrino-Mattos
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Camila S. de Figueiredo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Ícaro Raony
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (Í.R.); (H.R.F.)
| | - Aline Araujo dos Santos
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Aline Teixeira Duarte-Silva
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Mariana Santana Dias
- Laboratory of Gene Therapy and Viral Vectors, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.S.D.); (H.P.S.)
| | - Hércules Rezende Freitas
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (Í.R.); (H.R.F.)
| | - Elisabeth G. de Araújo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Victor Tulio Ribeiro-Resende
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Marcelo Cossenza
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Hilda P. Silva
- Laboratory of Gene Therapy and Viral Vectors, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.S.D.); (H.P.S.)
| | - Roberto P. de Carvalho
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Ana L. M. Ventura
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Karin C. Calaza
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Mariana S. Silveira
- Laboratory for Investigation in Neuroregeneration and Development, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil;
| | - Regina C. C. Kubrusly
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Ricardo A. de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| |
Collapse
|
6
|
Salazar K, Jara N, Ramírez E, de Lima I, Smith-Ghigliotto J, Muñoz V, Ferrada L, Nualart F. Role of vitamin C and SVCT2 in neurogenesis. Front Neurosci 2023; 17:1155758. [PMID: 37424994 PMCID: PMC10324519 DOI: 10.3389/fnins.2023.1155758] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Different studies have established the fundamental role of vitamin C in proliferation, differentiation, and neurogenesis in embryonic and adult brains, as well as in in vitro cell models. To fulfill these functions, the cells of the nervous system regulate the expression and sorting of sodium-dependent vitamin C transporter 2 (SVCT2), as well as the recycling of vitamin C between ascorbic acid (AA) and dehydroascorbic acid (DHA) via a bystander effect. SVCT2 is a transporter preferentially expressed in neurons and in neural precursor cells. In developmental stages, it is concentrated in the apical region of the radial glia, and in adult life, it is expressed preferentially in motor neurons of the cerebral cortex, starting on postnatal day 1. In neurogenic niches, SVCT2 is preferentially expressed in precursors with intermediate proliferation, where a scorbutic condition reduces neuronal differentiation. Vitamin C is a potent epigenetic regulator in stem cells; thus, it can induce the demethylation of DNA and histone H3K27m3 in the promoter region of genes involved in neurogenesis and differentiation, an effect mediated by Tet1 and Jmjd3 demethylases, respectively. In parallel, it has been shown that vitamin C induces the expression of stem cell-specific microRNA, including the Dlk1-Dio3 imprinting region and miR-143, which promotes stem cell self-renewal and suppresses de novo expression of the methyltransferase gene Dnmt3a. The epigenetic action of vitamin C has also been evaluated during gene reprogramming of human fibroblasts to induced pluripotent cells, where it has been shown that vitamin C substantially improves the efficiency and quality of reprogrammed cells. Thus, for a proper effect of vitamin C on neurogenesis and differentiation, its function as an enzymatic cofactor, modulator of gene expression and antioxidant is essential, as is proper recycling from DHA to AA by various supporting cells in the CNS.
Collapse
Affiliation(s)
- Katterine Salazar
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Center for Advanced Microscopy CMA BIO, University of Concepcion, Concepcion, Chile
| | - Nery Jara
- Department of Pharmacology, University of Concepcion, Concepcion, Chile
| | - Eder Ramírez
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Isabelle de Lima
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Javiera Smith-Ghigliotto
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
| | - Valentina Muñoz
- Department of Pharmacology, University of Concepcion, Concepcion, Chile
| | - Luciano Ferrada
- Center for Advanced Microscopy CMA BIO, University of Concepcion, Concepcion, Chile
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile
- Center for Advanced Microscopy CMA BIO, University of Concepcion, Concepcion, Chile
| |
Collapse
|
7
|
Chu Z, Yang J, Zheng W, Sun J, Wang W, Qian H. Recent advances on modulation of H2O2 in tumor microenvironment for enhanced cancer therapeutic efficacy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
8
|
Bedhiafi T, Idoudi S, Fernandes Q, Al-Zaidan L, Uddin S, Dermime S, Billa N, Merhi M. Nano-vitamin C: A promising candidate for therapeutic applications. Biomed Pharmacother 2023; 158:114093. [PMID: 36495664 DOI: 10.1016/j.biopha.2022.114093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/27/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Vitamin C is an important nutrient implicated in different physiological functions in humans. Despite its important biological functions, therapeutic applications of vitamin C are rare and its use is further impacted by low chemical stability. Several nano-encapsulation techniques have been described in the literature and yet, there are only a handful of clinical investigations dedicated to unlocking the therapeutic applications of nano-encapsulated vitamin C. Clearly, further investigations are warranted in order to affirm the promising clinical potential of nano-encapsulated vitamin C. In this review, we describe the mechanisms of vitamin C activity as a modulator of crucial therapeutic uses in biological systems. We look at key factors affecting the chemical stability of vitamin C alone and in nano-encapsulated and explore pre-clinical and clinical evidence on current vitamin C nano-formulations along with their therapeutic applications. Finally, we critically appraise the gaps and opportunities prevailing in nano-vitamin C research and its potential translation towards relevant clinical outcomes.
Collapse
Affiliation(s)
- Takwa Bedhiafi
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | | | - Queenie Fernandes
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; College of Medicine, Qatar University, Doha, Qatar
| | - Lobna Al-Zaidan
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | | | - Maysaloun Merhi
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
9
|
Travaglini S, Gurnari C, Antonelli S, Silvestrini G, Noguera NI, Ottone T, Voso MT. The Anti-Leukemia Effect of Ascorbic Acid: From the Pro-Oxidant Potential to the Epigenetic Role in Acute Myeloid Leukemia. Front Cell Dev Biol 2022; 10:930205. [PMID: 35938170 PMCID: PMC9352950 DOI: 10.3389/fcell.2022.930205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Data derived from high-throughput sequencing technologies have allowed a deeper understanding of the molecular landscape of Acute Myeloid Leukemia (AML), paving the way for the development of novel therapeutic options, with a higher efficacy and a lower toxicity than conventional chemotherapy. In the antileukemia drug development scenario, ascorbic acid, a natural compound also known as Vitamin C, has emerged for its potential anti-proliferative and pro-apoptotic activities on leukemic cells. However, the role of ascorbic acid (vitamin C) in the treatment of AML has been debated for decades. Mechanistic insight into its role in many biological processes and, especially, in epigenetic regulation has provided the rationale for the use of this agent as a novel anti-leukemia therapy in AML. Acting as a co-factor for 2-oxoglutarate-dependent dioxygenases (2-OGDDs), ascorbic acid is involved in the epigenetic regulations through the control of TET (ten-eleven translocation) enzymes, epigenetic master regulators with a critical role in aberrant hematopoiesis and leukemogenesis. In line with this discovery, great interest has been emerging for the clinical testing of this drug targeting leukemia epigenome. Besides its role in epigenetics, ascorbic acid is also a pivotal regulator of many physiological processes in human, particularly in the antioxidant cellular response, being able to scavenge reactive oxygen species (ROS) to prevent DNA damage and other effects involved in cancer transformation. Thus, for this wide spectrum of biological activities, ascorbic acid possesses some pharmacologic properties attractive for anti-leukemia therapy. The present review outlines the evidence and mechanism of ascorbic acid in leukemogenesis and its therapeutic potential in AML. With the growing evidence derived from the literature on situations in which the use of ascorbate may be beneficial in vitro and in vivo, we will finally discuss how these insights could be included into the rational design of future clinical trials.
Collapse
Affiliation(s)
- S. Travaglini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - C. Gurnari
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - S. Antonelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - G. Silvestrini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - N. I. Noguera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - T. Ottone
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - M. T. Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
- *Correspondence: M. T. Voso,
| |
Collapse
|
10
|
Opioids and Vitamin C: Known Interactions and Potential for Redox-Signaling Crosstalk. Antioxidants (Basel) 2022; 11:antiox11071267. [PMID: 35883757 PMCID: PMC9312198 DOI: 10.3390/antiox11071267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 12/10/2022] Open
Abstract
Opioids are among the most widely used classes of pharmacologically active compounds both clinically and recreationally. Beyond their analgesic efficacy via μ opioid receptor (MOR) agonism, a prominent side effect is central respiratory depression, leading to systemic hypoxia and free radical generation. Vitamin C (ascorbic acid; AA) is an essential antioxidant vitamin and is involved in the recycling of redox cofactors associated with inflammation. While AA has been shown to reduce some of the negative side effects of opioids, the underlying mechanisms have not been explored. The present review seeks to provide a signaling framework under which MOR activation and AA may interact. AA can directly quench reactive oxygen and nitrogen species induced by opioids, yet this activity alone does not sufficiently describe observations. Downstream of MOR activation, confounding effects from AA with STAT3, HIF1α, and NF-κB have the potential to block production of antioxidant proteins such as nitric oxide synthase and superoxide dismutase. Further mechanistic research is necessary to understand the underlying signaling crosstalk of MOR activation and AA in the amelioration of the negative, potentially fatal side effects of opioids.
Collapse
|
11
|
Ottone T, Faraoni I, Fucci G, Divona M, Travaglini S, De Bellis E, Marchesi F, Angelini DF, Palmieri R, Gurnari C, Giansanti M, Nardozza AM, Montesano F, Fabiani E, Lindfors Rossi EL, Cerretti R, Cicconi L, De Bardi M, Catanoso ML, Battistini L, Massoud R, Venditti A, Voso MT. Vitamin C Deficiency in Patients With Acute Myeloid Leukemia. Front Oncol 2022; 12:890344. [PMID: 35832559 PMCID: PMC9271703 DOI: 10.3389/fonc.2022.890344] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Vitamin C has been shown to play a significant role in suppressing progression of leukemia through epigenetic mechanisms. We aimed to study the role of vitamin C in acute myeloid leukemia (AML) biology and clinical course. To this purpose, the plasma levels of vitamin C at diagnosis in 62 patients with AML (including 5 cases with acute promyelocytic leukemia, APL),7 with myelodysplastic syndrome (MDS), and in 15 healthy donors (HDs) were studied. As controls, vitamins A and E levels were analysed. Expression of the main vitamin C transporters and of the TET2 enzyme were investigated by a specific RQ-PCR while cytoplasmic vitamin C concentration and its uptake were studied in mononuclear cells (MNCs), lymphocytes and blast cells purified from AML samples, and MNCs isolated from HDs. There were no significant differences in vitamin A and E serum levels between patients and HDs. Conversely, vitamin C concentration was significantly lower in AML as compared to HDs (p<0.0001), inversely correlated with peripheral blast‐counts (p=0.029), significantly increased at the time of complete remission (CR) (p=0.04) and further decreased in resistant disease (p=0.002). Expression of the main vitamin C transporters SLC23A2, SLC2A1 and SLC2A3 was also significantly reduced in AML compared to HDs. In this line, cytoplasmic vitamin C levels were also significantly lower in AML-MNCs versus HDs, and in sorted blasts compared to normal lymphocytes in individual patients. No association was found between vitamin C plasma levels and the mutation profile of AML patients, as well as when considering cytogenetics or 2017 ELN risk stratification groups. Finally, vitamin C levels did not play a predictive role for overall or relapse-free survival. In conclusion, our study shows that vitamin C levels are significantly decreased in patients with AML at the time of initial diagnosis, further decrease during disease progression and return to normal upon achievement of CR. Correspondingly, low intracellular levels may mirror increased vitamin C metabolic consumption in proliferating AML cells.
Collapse
Affiliation(s)
- Tiziana Ottone
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuro-Oncohematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
- *Correspondence: Tiziana Ottone,
| | - Isabella Faraoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giorgio Fucci
- Department of Experimental Medicine and Surgery, Faculty of Medicine and Surgery, University Tor Vergata, Rome, Italy
| | - Mariadomenica Divona
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- UniCamillus‐Saint Camillus International University of Health Sciences, Rome, Italy
| | - Serena Travaglini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Eleonora De Bellis
- Department of Biomedicine and Prevention, PhD in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
- Struttura Complessa (SC) Ematologia, Azienda Sanitaria Universitaria Giuliano Isontina Trieste, Trieste, Italy
| | - Francesco Marchesi
- Hematology and Stem Cell Transplant Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Regina Elena National Cancer Institute, Rome, Italy
| | - Daniela Francesca Angelini
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Raffaele Palmieri
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Carmelo Gurnari
- Department of Biomedicine and Prevention, PhD in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Manuela Giansanti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Anna Maria Nardozza
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Federica Montesano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Emiliano Fabiani
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- UniCamillus‐Saint Camillus International University of Health Sciences, Rome, Italy
| | | | - Raffaella Cerretti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Laura Cicconi
- Ospedale Santo Spirito, Azienda Sanitaria Locale (ASL) Roma 1, Reparto di Ematologia, Rome, Italy
| | - Marco De Bardi
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Maria Luisa Catanoso
- Department of Biomedicine and Prevention, PhD in Immunology, Molecular Medicine and Applied Biotechnology, University of Rome Tor Vergata, Rome, Italy
- Department Hematology/Oncology and Cell and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesú, Rome, Italy
| | - Luca Battistini
- Neuroimmunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Renato Massoud
- Department of Experimental Medicine and Surgery, Faculty of Medicine and Surgery, University Tor Vergata, Rome, Italy
| | - Adriano Venditti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuro-Oncohematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
12
|
High-Dose Vitamin C Supplementation as a Legitimate Anti-SARS-CoV-2 Prophylaxis in Healthy Subjects—Yes or No? Nutrients 2022; 14:nu14050979. [PMID: 35267953 PMCID: PMC8912816 DOI: 10.3390/nu14050979] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/13/2022] Open
Abstract
Vitamin C has a number of acitvities that could contribute to its immune-modulating effects. The only question is whether we should provide ourselves with only the right level of it, or do we need much more during a pandemic? The possibility of reducing the incidence of viral diseases in a well-nourished population through the use of dietary supplements based on vitamin C is not supported in the literature. Despite this, the belief that an extra intake of vitamin C can increase the efficacy of the immune system is still popular and vitamin C is advertised as a remedy to prevent infectious disease. This article refers to the justification of the use of vitamin C in high doses as an anti-SARS-CoV-2 prophylaxis in healthy subjects. Does it make sense or not? As it turns out, any effects of vitamin C supplementation may be more prominent when the baseline vitamin C level is low, for example in physically active persons. People with hypovitaminosis C are more likely to respond to vitamin C administration. No studies regarding prevention of COVID-19 with high-dose vitamin C supplementation in healthy subjects were found.
Collapse
|
13
|
Antioxidative Reactivity of L-Ascorbic Acid and D-Isoascorbic Acid Species towards Reduction of Hexachloroiridate (IV). J CHEM-NY 2021. [DOI: 10.1155/2021/5505741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The pair [IrCl6]2–/[IrCl6]3– has been demonstrated to be a good redox probe in biological systems while L-ascorbic acid (AA) is one of the most important antioxidants. D-isoascorbic acid (IAA) is an epimer of AA and is widely used as an antioxidant in various foods, beverages, meat, and fisher products. Reductions of [IrCl6]2– by AA and IAA have been analyzed kinetically and mechanistically in this work. The reductions strictly follow overall second-order kinetics and the observed second-order rate constants were collected in the pH region of 0 ≤ pH ≤ 2.33 at 25.0°C. Spectrophotometric titration experiments revealed a well-defined 1 : 2 stoichiometry, namely Δ[AA] : Δ[Ir(IV)] or Δ[IAA] : Δ[Ir(IV)] = 1 : 2, indicating that L-dehydroascorbic acid (DHA) and D-dehydroisoascorbic acid (DHIA) were the oxidation products of AA and IAA, respectively. A reaction mechanism is suggested involving parallel reactions of [IrCl6]2– with three protolysis species of AA/IAA (fully protonated, monoanionic, and dianionic forms) as the rate-determining steps and formation of ascorbic/isoascorbic and ascorbate/isoascorbate radicals; in each of the steps, [IrCl6]2– acquires an electron via an outer-sphere electron transfer mode. Rate constants of the rate-determining steps have been derived or estimated. The fully protonated forms of AA and IAA display virtually identical reactivity whereas ascorbate and isoascorbate monoanions have a significant reactivity difference. The ascorbate and isoascorbate dianions are extremely reactive and their reactions with [IrCl6]2– proceed with the diffusion-controlled rate. The species versus pH and the species reactivity versus pH distribution diagrams were constructed endowing that the ascorbate/isoascorbate monoanionic form dominated the total reactivity at physiological pH. In addition, the value of pKa1 = 3.74 ± 0.05 for IAA at 25.0°C and 1.0 M ionic strength was determined in this work.
Collapse
|
14
|
SVCT2 Overexpression and Ascorbic Acid Uptake Increase Cortical Neuron Differentiation, Which Is Dependent on Vitamin C Recycling between Neurons and Astrocytes. Antioxidants (Basel) 2021; 10:antiox10091413. [PMID: 34573045 PMCID: PMC8465431 DOI: 10.3390/antiox10091413] [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: 08/01/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Abstract
During brain development, sodium–vitamin C transporter (SVCT2) has been detected primarily in radial glial cells in situ, with low-to-absent expression in cerebral cortex neuroblasts. However, strong SVCT2 expression is observed during the first postnatal days, resulting in increased intracellular concentration of vitamin C. Hippocampal neurons isolated from SVCT2 knockout mice showed shorter neurites and low clustering of glutamate receptors. Other studies have shown that vitamin C-deprived guinea pigs have reduced spatial memory, suggesting that ascorbic acid (AA) and SVCT2 have important roles in postnatal neuronal differentiation and neurite formation. In this study, SVCT2 lentiviral overexpression induced branching and increased synaptic proteins expression in primary cultures of cortical neurons. Analysis in neuroblastoma 2a (Neuro2a) and human subventricular tumor C3 (HSVT-C3) cells showed similar branching results. SVCT2 was mainly observed in the cell membrane and endoplasmic reticulum; however, it was not detected in the mitochondria. Cellular branching in neuronal cells and in a previously standardized neurosphere assay is dependent on the recycling of vitamin C or reduction in dehydroascorbic acid (DHA, produced by neurons) by glial cells. The effect of WZB117, a selective glucose/DHA transporter 1 (GLUT1) inhibitor expressed in glial cells, was also studied. By inhibiting GLUT1 glial cells, a loss of branching is observed in vitro, which is reproduced in the cerebral cortex in situ. We concluded that vitamin C recycling between neurons and astrocyte-like cells is fundamental to maintain neuronal differentiation in vitro and in vivo. The recycling activity begins at the cerebral postnatal cortex when neurons increase SVCT2 expression and concomitantly, GLUT1 is expressed in glial cells.
Collapse
|