1
|
Whitsitt Q, Saxena A, Patel B, Evans BM, Hunt B, Purcell EK. Spatial transcriptomics at the brain-electrode interface in rat motor cortex and the relationship to recording quality. J Neural Eng 2024; 21:046033. [PMID: 38885679 PMCID: PMC11289622 DOI: 10.1088/1741-2552/ad5936] [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: 11/17/2023] [Revised: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024]
Abstract
Study of the foreign body reaction to implanted electrodes in the brain is an important area of research for the future development of neuroprostheses and experimental electrophysiology. After electrode implantation in the brain, microglial activation, reactive astrogliosis, and neuronal cell death create an environment immediately surrounding the electrode that is significantly altered from its homeostatic state.Objective.To uncover physiological changes potentially affecting device function and longevity, spatial transcriptomics (ST) was implemented to identify changes in gene expression driven by electrode implantation and compare this differential gene expression to traditional metrics of glial reactivity, neuronal loss, and electrophysiological recording quality.Approach.For these experiments, rats were chronically implanted with functional Michigan-style microelectrode arrays, from which electrophysiological recordings (multi-unit activity, local field potential) were taken over a six-week time course. Brain tissue cryosections surrounding each electrode were then mounted for ST processing. The tissue was immunolabeled for neurons and astrocytes, which provided both a spatial reference for ST and a quantitative measure of glial fibrillary acidic protein and neuronal nuclei immunolabeling surrounding each implant.Main results. Results from rat motor cortex within 300µm of the implanted electrodes at 24 h, 1 week, and 6 weeks post-implantation showed up to 553 significantly differentially expressed (DE) genes between implanted and non-implanted tissue sections. Regression on the significant DE genes identified the 6-7 genes that had the strongest relationship to histological and electrophysiological metrics, revealing potential candidate biomarkers of recording quality and the tissue response to implanted electrodes.Significance. Our analysis has shed new light onto the potential mechanisms involved in the tissue response to implanted electrodes while generating hypotheses regarding potential biomarkers related to recorded signal quality. A new approach has been developed to understand the tissue response to electrodes implanted in the brain using genes identified through transcriptomics, and to screen those results for potential relationships with functional outcomes.
Collapse
Affiliation(s)
- Quentin Whitsitt
- Department of Biomedical Engineering and Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Akash Saxena
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Bella Patel
- Department of Biomedical Engineering and Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Blake M Evans
- Department of Biomedical Engineering and Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Bradley Hunt
- Department of Biomedical Engineering and Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Erin K Purcell
- Department of Biomedical Engineering and Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| |
Collapse
|
2
|
Scibetta S, Miceli M, Iuliano M, Stefanuto L, Carbone E, Piscopo P, Petrozza V, Romeo G, Mangino G, Calogero A, Gasperi T, Rosa P. In Vitro Evaluation of the Antioxidant Capacity of 3,3-Disubstituted-3H-benzofuran-2-one Derivatives in a Cellular Model of Neurodegeneration. Life (Basel) 2024; 14:422. [PMID: 38672695 PMCID: PMC11051253 DOI: 10.3390/life14040422] [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: 02/14/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Oxidative stress represents a hallmark for many degenerative pathologies of the Central Nervous System. Throughout life, the constant pressure of noxious stimuli and/or episodes of traumatic events may expose the brain to a microenvironment where the non-balanced reactive oxygen species inevitably lead to neuronal loss and cognitive decline. HO-1, a 32 kDa heat-shock protein catalyzing the degradation of heme into carbon monoxide (CO), iron and biliverdin/bilirubin is considered one of the main antioxidant defense mechanisms playing pivotal roles in neuroprotection. Restoring the redox homeostasis is the goal of many natural or synthetic antioxidant molecules pursuing beneficial effects on brain functions. Here, we investigated the antioxidant capacity of four selected benzofuran-2-one derivatives in a cellular model of neurodegeneration represented by differentiated SH-SY5Y cells exposed to catechol-induced oxidative stress. Our main results highlight how all the molecules have antioxidant properties, especially compound 9, showing great abilities in reducing intracellular ROS levels and protecting differentiated SH-SY5Y cells from catechol-induced death. This compound above all seems to boost HO-1 mRNA and perinuclear HO-1 protein isoform expression when cells are exposed to the oxidative insult. Our findings open the way to consider benzofuran-2-ones as a novel and promising adjuvant antioxidant strategy for many neurodegenerative disorders.
Collapse
Affiliation(s)
- Sofia Scibetta
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
| | - Martina Miceli
- Department of Science, University of Roma Tre, 00146 Rome, Italy; (M.M.); (L.S.)
| | - Marco Iuliano
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
| | - Luca Stefanuto
- Department of Science, University of Roma Tre, 00146 Rome, Italy; (M.M.); (L.S.)
| | - Elena Carbone
- Department of Neuroscience, Italian National Institute of Health, 00161 Rome, Italy; (E.C.); (P.P.)
| | - Paola Piscopo
- Department of Neuroscience, Italian National Institute of Health, 00161 Rome, Italy; (E.C.); (P.P.)
| | - Vincenzo Petrozza
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
- Istituto Chirurgico Ortopedico Traumatologico (ICOT), 04100 Latina, Italy
| | - Giovanna Romeo
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
| | - Giorgio Mangino
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
- Istituto Chirurgico Ortopedico Traumatologico (ICOT), 04100 Latina, Italy
| | - Tecla Gasperi
- Department of Science, University of Roma Tre, 00146 Rome, Italy; (M.M.); (L.S.)
- National Institute of Biostructures and Biosystems (INBB), 00136 Rome, Italy
| | - Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
- Istituto Chirurgico Ortopedico Traumatologico (ICOT), 04100 Latina, Italy
| |
Collapse
|
3
|
Andrés CMC, Pérez de la Lastra JM, Juan CA, Plou FJ, Pérez-Lebeña E. Antioxidant Metabolism Pathways in Vitamins, Polyphenols, and Selenium: Parallels and Divergences. Int J Mol Sci 2024; 25:2600. [PMID: 38473850 DOI: 10.3390/ijms25052600] [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: 11/26/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Free radicals (FRs) are unstable molecules that cause reactive stress (RS), an imbalance between reactive oxygen and nitrogen species in the body and its ability to neutralize them. These species are generated by both internal and external factors and can damage cellular lipids, proteins, and DNA. Antioxidants prevent or slow down the oxidation process by interrupting the transfer of electrons between substances and reactive agents. This is particularly important at the cellular level because oxidation reactions lead to the formation of FR and contribute to various diseases. As we age, RS accumulates and leads to organ dysfunction and age-related disorders. Polyphenols; vitamins A, C, and E; and selenoproteins possess antioxidant properties and may have a role in preventing and treating certain human diseases associated with RS. In this review, we explore the current evidence on the potential benefits of dietary supplementation and investigate the intricate connection between SIRT1, a crucial regulator of aging and longevity; the transcription factor NRF2; and polyphenols, vitamins, and selenium. Finally, we discuss the positive effects of antioxidant molecules, such as reducing RS, and their potential in slowing down several diseases.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain
| | - Francisco J Plou
- Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, 28049 Madrid, Spain
| | | |
Collapse
|
4
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
5
|
Hyponatremia and Cancer: From Bedside to Benchside. Cancers (Basel) 2023; 15:cancers15041197. [PMID: 36831539 PMCID: PMC9953859 DOI: 10.3390/cancers15041197] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Hyponatremia is the most common electrolyte disorder encountered in hospitalized patients. This applies also to cancer patients. Multiple causes can lead to hyponatremia, but most frequently this electrolyte disorder is due to the syndrome of inappropriate antidiuresis. In cancer patients, this syndrome is mostly secondary to ectopic secretion of arginine vasopressin by tumoral cells. In addition, several chemotherapeutic drugs induce the release of arginine vasopressin by the hypothalamus. There is evidence that hyponatremia is associated to a more negative outcome in several pathologies, including cancer. Many studies have demonstrated that in different cancer types, both progression-free survival and overall survival are negatively affected by hyponatremia, whereas the correction of serum [Na+] has a positive effect on patient outcome. In vitro studies have shown that cells grown in low [Na+] have a greater proliferation rate and motility, due to a dysregulation in intracellular signalling pathways. Noteworthy, vasopressin receptors antagonists, which were approved more than a decade ago for the treatment of euvolemic and hypervolemic hyponatremia, have shown unexpected antiproliferative effects. Because of this property, vaptans were also approved for the treatment of polycystic kidney disease. In vitro evidence indicated that this family of drugs effectively counteracts proliferation and invasivity of cancer cells, thus possibly opening a new scenario among the pharmacological strategies to treat cancer.
Collapse
|
6
|
Mansor NI, Ling KH, Rosli R, Hassan Z, Adenan MI, Nordin N. Centella asiatica (L.) Urban. Attenuates Cell Damage in Hydrogen Peroxide-Induced Oxidative Stress in Transgenic Murine Embryonic Stem Cell Line-Derived Neural-Like Cells: A Preliminary Study for Potential Treatment of Alzheimer's Disease. J Alzheimers Dis 2023; 94:S21-S44. [PMID: 37334592 PMCID: PMC10473099 DOI: 10.3233/jad-221233] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Centella asiatica (L.) (C. asiatica) is commonly known in South East and South East Asia communities for its nutritional and medicinal benefits. Besides being traditionally used to enhance memory and accelerate wound healing, its phytochemicals have been extensively documented for their neuroprotective, neuroregenerative, and antioxidant properties. OBJECTIVE The present study aims to investigate the effects of a standardized raw extract of C. asiatica (RECA) on hydrogen peroxide (H2O2)-induced oxidative stress and apoptotic death in neural-like cells derived from mouse embryonic stem (ES) cell line. METHODS A transgenic mouse ES cell (46C) was differentiated into neural-like cells using 4-/4+ protocol with addition of all-trans retinoic acid. These cells were then exposed to H2O2 for 24 h. The effects of RECA on H2O2-induced neural-like cells were assessed through cell viability, apoptosis, and reactive oxygen species (ROS) assays, as well as neurite length measurement. The gene expression levels of neuronal-specific and antioxidant markers were assessed by RT-qPCR analysis. RESULTS Pre-treatment with H2O2 for 24 hours, in a dose-dependent manner, damaged neural-like cells as marked by a decrease in cell viability, substantial increase in intracellular ROS accumulation, and increase in apoptotic rate compared to untreated cells. These cells were used to treat with RECA. Treatment with RECA for 48 h remarkably restored cell survival and promoted neurite outgrowth in the H2O2- damaged neurons by increasing cell viability and decreasing ROS activity. RT-qPCR analysis revealed that RECA upregulated the level of antioxidant genes such as thioredoxin-1 (Trx-1) and heme oxygenase-1 (HO-1) of treated cells, as well as the expression level of neuronal-specific markers such as Tuj1 and MAP2 genes, suggesting their contribution in neuritogenic effect. CONCLUSION Our findings indicate that RECA promotes neuroregenerative effects and exhibits antioxidant properties, suggesting a valuable synergistic activity of its phytochemical constituents, thus, making the extract a promising candidate in preventing or treating oxidative stress-associated Alzheimer's disease.
Collapse
Affiliation(s)
- Nur Izzati Mansor
- Medical Genetics Unit, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Department of Nursing, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras Kuala Lumpur, Malaysia
| | - King-Hwa Ling
- Medical Genetics Unit, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Malaysian Research Institute on Ageing (MyAgeing™), Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine (ReGEN) Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Rozita Rosli
- Medical Genetics Unit, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine (ReGEN) Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Gelugor, Penang, Malaysia
| | - Mohd Ilham Adenan
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Universiti Teknologi MARA, Puncak Alam Campus, Bandar PuncakAlam, Selangor Darul Ehsan, Malaysia
| | - Norshariza Nordin
- Medical Genetics Unit, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Malaysian Research Institute on Ageing (MyAgeing™), Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine (ReGEN) Research Group, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
7
|
Agmatine-mediated inhibition of NMDA receptor expression and amelioration of dyskinesia via activation of Nrf2 and suppression of HMGB1/RAGE/TLR4/MYD88/NF-κB signaling cascade in rotenone lesioned rats. Life Sci 2022; 311:121049. [DOI: 10.1016/j.lfs.2022.121049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 11/19/2022]
|
8
|
Yin H, Chen Z, Zhao H, Huang H, Liu W. Noble gas and neuroprotection: From bench to bedside. Front Pharmacol 2022; 13:1028688. [PMID: 36532733 PMCID: PMC9750501 DOI: 10.3389/fphar.2022.1028688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/03/2022] [Indexed: 07/26/2023] Open
Abstract
In recent years, inert gases such as helium, argon, and xenon have gained considerable attention for their medical value. Noble gases present an intriguing scientific paradox: although extremely chemically inert, they display a remarkable spectrum of clinically useful biological properties. Despite a relative paucity of knowledge about their mechanisms of action, some noble gases have been used successfully in clinical practice. The neuroprotection elicited by these noble gases has been investigated in experimental animal models of various types of brain injuries, such as traumatic brain injury, stroke, subarachnoid hemorrhage, cerebral ischemic/reperfusion injury, and neurodegenerative diseases. Collectively, these central nervous system injuries are a leading cause of morbidity and mortality every year worldwide. Treatment options are presently limited to thrombolytic drugs and clot removal for ischemic stroke, or therapeutic cooling for other brain injuries before the application of noble gas. Currently, there is increasing interest in noble gases as novel treatments for various brain injuries. In recent years, neuroprotection elicited by particular noble gases, xenon, for example, has been reported under different conditions. In this article, we have reviewed the latest in vitro and in vivo experimental and clinical studies of the actions of xenon, argon, and helium, and discuss their potential use as neuroprotective agents.
Collapse
Affiliation(s)
- Haiying Yin
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zijun Chen
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hailin Zhao
- Division of Anesthetics, Department of Surgery and Cancer, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Han Huang
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wenwen Liu
- Department of Anesthesia Nursing, West China Second University Hospital, Sichuan University/West China School of Nursing, Ministry of Education, Sichuan University and Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, China
| |
Collapse
|
9
|
Dihydromyricetin ameliorates social isolation-induced anxiety by modulating mitochondrial function, antioxidant enzymes, and BDNF. Neurobiol Stress 2022; 21:100499. [DOI: 10.1016/j.ynstr.2022.100499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 11/08/2022] Open
|
10
|
Cressatti M, Schipper HM. Dysregulation of a Heme Oxygenase-Synuclein Axis in Parkinson Disease. NEUROSCI 2022; 3:284-299. [PMID: 39483365 PMCID: PMC11523740 DOI: 10.3390/neurosci3020020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 11/03/2024] Open
Abstract
α-Synuclein is a key driver of the pathogenesis of Parkinson disease (PD). Heme oxygenase-1 (HO-1), a stress protein that catalyzes the conversion of heme to biliverdin, carbon monoxide and free ferrous iron, is elevated in PD-affected neural tissues and promotes iron deposition and mitochondrial dysfunction in models of the disease, pathways also impacted by α-synuclein. Elevated expression of human HO-1 in astrocytes of GFAP.HMOX1 transgenic mice between 8.5 and 19 months of age elicits a parkinsonian phenotype characterized by nigrostriatal hypodopaminergia, locomotor incoordination and overproduction of neurotoxic native S129-phospho-α-synuclein. Two microRNAs (miRNA) known to regulate α-synuclein, miR-153 and miR-223, are significantly decreased in the basal ganglia of GFAP.HMOX1 mice. Serum concentrations of both miRNAs progressively decline in wild-type (WT) and GFAP.HMOX1 mice between 11 and 18 months of age. Moreover, circulating levels of miR-153 and miR-223 are significantly lower, and erythrocyte α-synuclein concentrations are increased, in GFAP.HMOX1 mice relative to WT values. MiR-153 and miR-223 are similarly decreased in the saliva of PD patients compared to healthy controls. Upregulation of glial HO-1 may promote parkinsonism by suppressing miR-153 and miR-223, which, in turn, enhance production of neurotoxic α-synuclein. The aim of the current review is to explore the link between HO-1, α-synuclein and PD, evaluating evidence derived from our laboratory and others. HO-1, miR-153 and miR-223 and α-synuclein may serve as potential biomarkers and targets for disease-modifying therapy in idiopathic PD.
Collapse
Affiliation(s)
- Marisa Cressatti
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3T1E2, Canada;
- Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Neurology & Neurosurgery, McGill University, 3999 Cote Sainte-Catherine Road, Montreal, QC H3T1E2, Canada
| | - Hyman M Schipper
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3T1E2, Canada;
- Lady Davis Institute for Medical Research, Jewish General Hospital, Department of Neurology & Neurosurgery, McGill University, 3999 Cote Sainte-Catherine Road, Montreal, QC H3T1E2, Canada
| |
Collapse
|
11
|
Physical-Exercise-Induced Antioxidant Effects on the Brain and Skeletal Muscle. Antioxidants (Basel) 2022; 11:antiox11050826. [PMID: 35624690 PMCID: PMC9138070 DOI: 10.3390/antiox11050826] [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: 03/31/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Erythroid-related nuclear factor 2 (NRF2) and the antioxidant-responsive-elements (ARE) signaling pathway are the master regulators of cell antioxidant defenses, playing a key role in maintaining cellular homeostasis, a scenario in which proper mitochondrial function is essential. Increasing evidence indicates that the regular practice of physical exercise increases cellular antioxidant defenses by activating NRF2 signaling. This manuscript reviewed classic and ongoing research on the beneficial effects of exercise on the antioxidant system in both the brain and skeletal muscle.
Collapse
|
12
|
Lee H, Liu Z, Dong L, Cheong SH, Lee DS. Lycopus maackianus Makino MeOH Extract Exhibits Antioxidant and Anti-Neuroinflammatory Effects in Neuronal Cells and Zebrafish Model. Antioxidants (Basel) 2022; 11:antiox11040690. [PMID: 35453375 PMCID: PMC9025111 DOI: 10.3390/antiox11040690] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 11/18/2022] Open
Abstract
Lycopus maackianus Makino belongs to the Labiatae family and is used in traditional medicine to manage postpartum edema and boils. However, few studies on its antioxidant and anti-inflammatory effects have been conducted. Here, the compounds in L. maackianus methanol (MeOH) extract were profiled using ultra-high-performance liquid chromatography–time-of-flight high-resolution mass spectrometry analysis. The antioxidant activity of L. maackianus MeOH extract was shown to increase in a concentration-dependent manner by investigating the 2,2-diphenyl-1-picrylhydrazyl and 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) radical scavenging activity. Next, in lipopolysaccharide-treated BV2 cells, L. maackianus extract inactivated the nuclear factor-kappa B pathway, inhibiting nitric oxide, prostaglandin E2, interleukin-6, and tumor necrosis factor-α production and inducible nitric oxide synthase and cyclooxygenase-2 protein expression. Furthermore, L. maackianus extract protected against oxidative stress-induced cellular damage in glutamate-stimulated HT22 cells. L. maackianus MeOH extract induced heme oxygenase-1 expression and increased the translocation of nuclear factor E2-related factor 2 in the nucleus, thus exhibiting antioxidant and anti-inflammatory effects. Moreover, the in vivo antioxidant and anti-inflammatory effects of the extract were demonstrated in a zebrafish (Danio rerio) model treated with hydrogen peroxide and lipopolysaccharide. MeOH L. maackianus extract showed antioxidant and anti-neuroinflammatory effects by increasing the expression of heme oxygenase-1, establishing its therapeutic potential for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Hwan Lee
- College of Pharmacy, Chosun University, Dong-gu, Gwangju 61452, Korea; (H.L.); (Z.L.); (L.D.)
| | - Zhiming Liu
- College of Pharmacy, Chosun University, Dong-gu, Gwangju 61452, Korea; (H.L.); (Z.L.); (L.D.)
| | - Linsha Dong
- College of Pharmacy, Chosun University, Dong-gu, Gwangju 61452, Korea; (H.L.); (Z.L.); (L.D.)
| | - Sun Hee Cheong
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Korea;
| | - Dong-Sung Lee
- College of Pharmacy, Chosun University, Dong-gu, Gwangju 61452, Korea; (H.L.); (Z.L.); (L.D.)
- Correspondence: ; Tel.: +82-63-230-6386
| |
Collapse
|
13
|
Wang X, Wan M, Zhang L, Dai Y, Hai Y, Yue C, Xu J, Ding Y, Wang M, Xie J, Lei X, Zhong JL. ALA_PDT Promotes Ferroptosis-Like Death of Mycobacterium abscessus and Antibiotic Sterilization via Oxidative Stress. Antioxidants (Basel) 2022; 11:546. [PMID: 35326196 PMCID: PMC8945036 DOI: 10.3390/antiox11030546] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/06/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium abscessus is one of the common clinical non-tuberculous mycobacteria (NTM) that can cause severe skin infection. 5-Aminolevulinic acid photodynamic therapy (ALA_PDT) is an emerging effective antimicrobial treatment. To explore whether ALA_PDT can be used to treat M. abscessus infections, we conducted a series of experiments in vitro. We found that ALA_PDT can kill M. abscesses. Mechanistically, we found that ALA_PDT promoted ferroptosis-like death of M. abscesses, and the ROS scavenger N-Acetyl-L-cysteine (NAC) and ferroptosis inhibitor Ferrostatin-1 (Fer-1) can mitigate the ALA_PDT-mediated sterilization. Furthermore, ALA_PDT significantly up-regulated the transcription of heme oxygenase MAB_4773, increased the intracellular Fe2+ concentration and altered the transcription of M. abscessus iron metabolism genes. ALA_PDT disrupted the integrity of the cell membrane and enhanced the permeability of the cell membrane, as evidenced by the boosted sterilization effect of antibiotics. In summary, ALA_PDT can kill M. abscesses via promoting the ferroptosis-like death and antibiotic sterilization through oxidative stress by changing iron metabolism. The study provided new mechanistic insights into the clinical efficacy of ALA_PDT against M. abscessus.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (X.W.); (M.W.); (Y.D.); (M.W.)
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing 400044, China;
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, College of Life and Sciences, Southwest University, Chongqing 400700, China; (L.Z.); (Y.D.); (Y.H.); (J.X.)
| | - Meiyin Wan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (X.W.); (M.W.); (Y.D.); (M.W.)
| | - Lei Zhang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, College of Life and Sciences, Southwest University, Chongqing 400700, China; (L.Z.); (Y.D.); (Y.H.); (J.X.)
| | - Yongdong Dai
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, College of Life and Sciences, Southwest University, Chongqing 400700, China; (L.Z.); (Y.D.); (Y.H.); (J.X.)
| | - Yang Hai
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, College of Life and Sciences, Southwest University, Chongqing 400700, China; (L.Z.); (Y.D.); (Y.H.); (J.X.)
| | - Chenda Yue
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing 400044, China;
| | - Junqi Xu
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, College of Life and Sciences, Southwest University, Chongqing 400700, China; (L.Z.); (Y.D.); (Y.H.); (J.X.)
| | - Yadan Ding
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (X.W.); (M.W.); (Y.D.); (M.W.)
| | - Mei Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (X.W.); (M.W.); (Y.D.); (M.W.)
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, College of Life and Sciences, Southwest University, Chongqing 400700, China; (L.Z.); (Y.D.); (Y.H.); (J.X.)
| | - Xia Lei
- Department of Dermatology, Daping Hospital, The Army Medical University, Chongqing 400044, China;
| | - Julia-Li Zhong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; (X.W.); (M.W.); (Y.D.); (M.W.)
| |
Collapse
|
14
|
Ramya V, Shyam KP, Kowsalya E, Balavigneswaran CK, Kadalmani B. Dual Roles of Coconut Oil and Its Major Component Lauric Acid on Redox Nexus: Focus on Cytoprotection and Cancer Cell Death. Front Neurosci 2022; 16:833630. [PMID: 35360165 PMCID: PMC8963114 DOI: 10.3389/fnins.2022.833630] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/11/2022] [Indexed: 11/23/2022] Open
Abstract
It has been reported that coconut oil supplementation can reduce neuroinflammation. However, coconut oils are available as virgin coconut oil (VCO), crude coconut oil (ECO), and refined coconut oil (RCO). The impact of coconut oil extraction process (and its major fatty acid component lauric acid) at cellular antioxidant level, redox homeostasis and inflammation in neural cells is hitherto unexplained. Herein, we have shown the antioxidant levels and cellular effect of coconut oil extracted by various processes in human neuroblastoma cells (SH-SY5Y) cultured in vitro. Results indicate VCO and ECO treated cells displayed better mitochondrial health when compared to RCO. Similar trend was observed for the release of reactive oxygen species (ROS), key oxidative stress response genes (GCLC, HO-1, and Nqo1) and inflammatory genes (IL6, TNFα, and iNOS) in SH-SY5Y cells. Our results signified that both VCO and ECO offer better neural health primarily by maintaining the cellular redox balance. Further, RCO prepared by solvent extraction and chemical refining process lacks appreciable beneficial effect. Then, we extended our study to find out the reasons behind maintaining the cellular redox balance in neuroblastoma cells by VCO and ECO. Our GC-MS results showed that lauric acid (C14:0) (LA) content was the major difference in the fatty acid composition extracted by various processes. Therefore, we evaluated the efficacy of LA in SH-SY5Y cells. The LA showed dose-dependent effect. At IC50 concentration (11.8 μM), LA down regulated the oxidative stress response genes and inflammatory genes. The results clearly indicate that the LA inhibited the neuroinflammation and provided an efficient cellular antioxidant activity, which protects the cells. The efficiency was also evaluated in normal cell line such as fibroblasts (L929) to cross-validate that the results were not false positive. Different concentration of LA on L929 cells showed high compatibility. From our observation, we conclude that VCO and ECO offers better cellular protection owing to their powerful antioxidant system. Therefore, we advocate the inclusion of either VCO and/or ECO in the diet for a healthy lifestyle.
Collapse
Affiliation(s)
- Venkatesan Ramya
- Reproductive Endocrinology and Cancer Biology Laboratory, Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| | | | - Eshwaran Kowsalya
- Research and Development Division, V.V.D and Sons Private Limited, Thoothukudi, India
| | - Chelladurai Karthikeyan Balavigneswaran
- Tissue Engineering and Biomaterials Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Balamuthu Kadalmani
- Reproductive Endocrinology and Cancer Biology Laboratory, Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| |
Collapse
|
15
|
Abu-Elfotuh K, Al-Najjar AH, Mohammed AA, Aboutaleb AS, Badawi GA. Fluoxetine ameliorates Alzheimer's disease progression and prevents the exacerbation of cardiovascular dysfunction of socially isolated depressed rats through activation of Nrf2/HO-1 and hindering TLR4/NLRP3 inflammasome signaling pathway. Int Immunopharmacol 2022; 104:108488. [PMID: 35042170 DOI: 10.1016/j.intimp.2021.108488] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/29/2021] [Accepted: 12/16/2021] [Indexed: 12/11/2022]
Abstract
Depression is a risk factor for Alzheimer's (AD) and cardiovascular diseases (CVD). Therefore, depression treatment restricts its deteriorating effects on mood, memory and CV system. Fluoxetine is the most widely used antidepressant drug, it has neuroprotective effect through its antioxidant/anti-inflammatory properties. The current study investigated for the first-time the cross link between depression, AD and CVD besides, role of fluoxetine in mitigating such disorders. Depression was induced in rats by social isolation (SI) for 12 weeks, AlCL3 (70 mg/kg/day, i.p.) was used to induce AD which was administered either in SI or normal control (NC) grouped rats starting at 8th week till the end of the experiment, fluoxetine (10 mg/kg/day, p.o) treatment also was started at 8th week. SI and AD showed a statistically significant deteriorated effect on behavioral, neurochemical and histopathological analysis which was exaggerated when two disorder combined than each alone. Fluoxetine treatment showed protective effect against SI, AD and prevents exacerbation of CVD. Fluoxetine improved animals' behavior, increased brain monoamines, BDNF besides increased antioxidant defense mechanism of SOD, TAC contents and increased protein expression of Nrf2/HO-1 with significant decrease of AChE activity, β-amyloid, Tau protein, MDA, TNF-α, IL1β contents as well as decreased protein expression of NF-kB, TLR4, NLRP3 and caspase1. It also showed cardioprotective effects as it improved lipid profile with pronounced decrease of cardiac enzymes of CK-MB, troponin and MEF2. In conclusion, fluoxetine represents as a promising drug against central and peripheral disorders through its anti-inflammatory/antioxidant effects via targeting antioxidant Nrf2/HO-1 and hindering TLR4/NLRP3 inflammasome signaling pathways.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Aya H Al-Najjar
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Asmaa A Mohammed
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Amany S Aboutaleb
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Ghada A Badawi
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Sinai University, El Arish, Egypt.
| |
Collapse
|
16
|
Ren X, Zhang J, Zhao Y, Sun L. Senegenin Inhibits Aβ 1-42-Induced PC12 Cells Apoptosis and Oxidative Stress via Activation of the PI3K/Akt Signaling Pathway. Neuropsychiatr Dis Treat 2022; 18:513-524. [PMID: 35280979 PMCID: PMC8904946 DOI: 10.2147/ndt.s346238] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/10/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND/AIM Apoptosis and oxidative stress have been considered as key events in the pathogenesis of Alzheimer's disease (AD). Senegenin (Sen), the major and most effective ingredient of Radix Polygalae, which has anti-apoptotic and anti-oxidative effects. The aim of this study was to investigate the anti-apoptotic and anti-oxidant effects of Sen on Aβ1-42-induced PC12 cells apoptosis and oxidative stress as well as its possible signaling pathway. METHODS Rat pheochromocytoma (PC12) cells were treated by 20 μM Aβ1-42 and then divided into 5 different treatment groups (Control; Aβ1-42 20 μM; Aβ1-42 20 μM + Sen 10 μM; Aβ1-42 20 μM + Sen 30 μM; Aβ1-42 20μM + Sen 60 μM). PC12 cells activity was detected by MTT assay. Colony formation assay was performed to assess the clonogenic ability of cells. The cell apoptosis was detected by Annexin-V/PI staining. The pro-apoptotic protein (Bax), anti-apoptotic protein (Bcl-2), anti-oxidative stress factor (HO-1, Nuclear Nrf2, Total Nrf2) and pathway-related protein (Akt, P-Akt, PI3K, P-PI3K) were tested by Western blot. The reactive oxygen species (ROS) level was assessed with a DCFH-DA probe. RESULTS The results indicated that Sen dose-dependently increased cell viability and reduced the number of apoptotic cells. The ratio of P-PI3K/PI3K and P-Akt/Akt increased in a dose-dependent manner under the treatment of Sen, suggesting that Sen might activate the PI3K/Akt signaling pathway. Moreover, Sen upregulates the ratio of Bcl-2/Bax. Further study revealed that Sen can play an antioxidant role in enhancing HO-1, promoting Nrf2 nuclear translocation and reducing ROS accumulation to reduce oxidative stress. CONCLUSION Sen is effective in inhibiting apoptosis and oxidative stress in Aβ1-42-induced PC12 cells, which likely contribute to the development of novel therapies for AD.
Collapse
Affiliation(s)
- Xing Ren
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, People's Republic of China
| | - Jiwei Zhang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, 250014, People's Republic of China
| | - Yunnan Zhao
- Editorial Office of Journal of Shandong University of Traditional Chinese Medicine, Jinan, 250014, People's Republic of China
| | - Lingzhi Sun
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| |
Collapse
|
17
|
Walia V, Kaushik D, Mittal V, Kumar K, Verma R, Parashar J, Akter R, Rahman MH, Bhatia S, Al-Harrasi A, Karthika C, Bhattacharya T, Chopra H, Ashraf GM. Delineation of Neuroprotective Effects and Possible Benefits of AntioxidantsTherapy for the Treatment of Alzheimer's Diseases by Targeting Mitochondrial-Derived Reactive Oxygen Species: Bench to Bedside. Mol Neurobiol 2021; 59:657-680. [PMID: 34751889 DOI: 10.1007/s12035-021-02617-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/19/2021] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is considered the sixth leading cause of death in elderly patients and is characterized by progressive neuronal degeneration and impairment in memory, language, etc. AD is characterized by the deposition of senile plaque, accumulation of fibrils, and neurofibrillary tangles (NFTs) which are responsible for neuronal degeneration. Amyloid-β (Aβ) plays a key role in the process of neuronal degeneration in the case of AD. It has been reported that Aβ is responsible for the production of reactive oxygen species (ROS), depletion of endogenous antioxidants, increase in intracellular Ca2+ which further increases mitochondria dysfunctions, oxidative stress, release of pro-apoptotic factors, neuronal apoptosis, etc. Thus, oxidative stress plays a key role in the pathogenesis of AD. Antioxidants are compounds that have the ability to counteract the oxidative damage conferred by ROS. Therefore, the antioxidant therapy may provide benefits and halt the progress of AD to advance stages by counteracting neuronal degeneration. However, despite the beneficial effects imposed by the antioxidants, the findings from the clinical studies suggested inconsistent results which might be due to poor study design, selection of the wrong antioxidant, inability of the molecule to cross the blood-brain barrier (BBB), treatment in the advanced state of disease, etc. The present review insights into the neuroprotective effects and limitations of the antioxidant therapy for the treatment of AD by targeting mitochondrial-derived ROS. This particular article will certainly help the researchers to search new avenues for the treatment of AD by utilizing mitochondrial-derived ROS-targeted antioxidant therapies.
Collapse
Affiliation(s)
- Vaibhav Walia
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, India
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Kuldeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
- University Institute of Pharmaceutical Sciences (UIPS), Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Ravinder Verma
- Department of Pharmacy, School of Medical and Allied Sciences, G.D. Goenka University, Gurugram, 122103, India
| | - Jatin Parashar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka, 1100, Bangladesh
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka, 1213, Bangladesh.
| | - Saurabh Bhatia
- School of Health Science University of Petroleum and Energy Studies, Dehrandun, Uttarkhand, 248007, India
- Natural & Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mouz, P.O. Box 33, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, 616 Birkat Al Mouz, P.O. Box 33, Nizwa, Oman
| | - Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, The Nilgiris, Ooty, 643001, Tamil Nadu, India
| | - Tanima Bhattacharya
- College of Chemistry & Chemical Engineering, Hubei University, Wuhan, 430062, China
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
18
|
Fibbi B, Marroncini G, Anceschi C, Naldi L, Peri A. Hyponatremia and Oxidative Stress. Antioxidants (Basel) 2021; 10:1768. [PMID: 34829639 PMCID: PMC8614907 DOI: 10.3390/antiox10111768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/26/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022] Open
Abstract
Hyponatremia, i.e., the presence of a serum sodium concentration ([Na+]) < 136 mEq/L, is the most frequent electrolyte imbalance in the elderly and in hospitalized patients. Symptoms of acute hyponatremia, whose main target is the central nervous system, are explained by the "osmotic theory" and the neuronal swelling secondary to decreased extracellular osmolality, which determines cerebral oedema. Following the description of neurological and systemic manifestations even in mild and chronic hyponatremia, in the last decade reduced extracellular [Na+] was associated with detrimental effects on cellular homeostasis independently of hypoosmolality. Most of these alterations appeared to be elicited by oxidative stress. In this review, we focus on the role of oxidative stress on both osmolality-dependent and -independent impairment of cell and tissue functions observed in hyponatremic conditions. Furthermore, basic and clinical research suggested that oxidative stress appears to be a common denominator of the degenerative processes related to aging, cancer progression, and hyponatremia. Of note, low [Na+] is able to exacerbate multiple manifestations of senescence and to decrease progression-free and overall survival in oncologic patients.
Collapse
Affiliation(s)
- Benedetta Fibbi
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Giada Marroncini
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Cecilia Anceschi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Laura Naldi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Alessandro Peri
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| |
Collapse
|
19
|
Upadhayay S, Mehan S. Targeting Nrf2/HO-1 anti-oxidant signaling pathway in the progression of multiple sclerosis and influences on neurological dysfunctions. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
20
|
Caba E, Sherman MD, Farizatto KLG, Alcira B, Wang HW, Giardina C, Shin DG, Sandefur CI, Bahr BA. Excitotoxic stimulation activates distinct pathogenic and protective expression signatures in the hippocampus. J Cell Mol Med 2021; 25:9011-9027. [PMID: 34414662 PMCID: PMC8435451 DOI: 10.1111/jcmm.16864] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
Excitotoxic events underlying ischaemic and traumatic brain injuries activate degenerative and protective pathways, particularly in the hippocampus. To understand opposing pathways that determine the brain's response to excitotoxicity, we used hippocampal explants, thereby eliminating systemic variables during a precise protocol of excitatory stimulation. N‐methyl‐d‐aspartate (NMDA) was applied for 20 min and total RNA isolated one and 24 h later for neurobiology‐specific microarrays. Distinct groups of genes exhibited early vs. delayed induction, with 63 genes exclusively reduced 24‐h post‐insult. Egr‐1 and NOR‐1 displayed biphasic transcriptional modulation: early induction followed by delayed suppression. Opposing events of NMDA‐induced genes linked to pathogenesis and cell survival constituted the early expression signature. Delayed degenerative indicators (up‐regulated pathogenic genes, down‐regulated pro‐survival genes) and opposing compensatory responses (down‐regulated pathogenic genes, up‐regulated pro‐survival genes) generated networks with temporal gene profiles mirroring coexpression network clustering. We then used the expression profiles to test whether NF‐κB, a potent transcription factor implicated in both degenerative and protective pathways, is involved in the opposing responses. The NF‐κB inhibitor MG‐132 indeed altered NMDA‐mediated transcriptional changes, revealing components of opposing expression signatures that converge on the single response element. Overall, this study identified counteracting avenues among the distinct responses to excitotoxicity, thereby suggesting multi‐target treatment strategies and implications for predictive medicine.
Collapse
Affiliation(s)
- Ebru Caba
- Vertex Pharmaceuticals, Cambridge, MA, USA.,Department of Pharmaceutical Sciences and the Neurosciences Program, University of Connecticut, Storrs, CT, USA
| | - Marcus D Sherman
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Britney Alcira
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Hsin-Wei Wang
- Bioinformatics and Biocomputing Institute, University of Connecticut, Storrs, CT, USA.,Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Dong-Guk Shin
- Bioinformatics and Biocomputing Institute, University of Connecticut, Storrs, CT, USA.,Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Conner I Sandefur
- Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Pharmacology and the Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA.,Sandefur Modeling, Pittsboro, NC, USA
| | - Ben A Bahr
- Department of Pharmaceutical Sciences and the Neurosciences Program, University of Connecticut, Storrs, CT, USA.,Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Chemistry and Physics, University of North Carolina-Pembroke, Pembroke, NC, USA
| |
Collapse
|
21
|
Che J, Yang J, Zhao B, Shang P. HO-1: A new potential therapeutic target to combat osteoporosis. Eur J Pharmacol 2021; 906:174219. [PMID: 34081904 DOI: 10.1016/j.ejphar.2021.174219] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023]
Abstract
Heme oxygenase-1 (HO-1) exerts a protective effect against cell damage and induces the activity of many enzymes involved in the treatment of many human diseases, including osteoporosis. The increasing prevalence of osteoporosis and the limitations of the current treatments available led to a continuous occurrence of bone loss and osteoporotic fractures, highlighting the need of a better understanding of the mechanism and function of HO-1. Many factors cause osteoporosis, including lack of estrogen, aging, and iron overload, and they either cause the increase in inflammatory factors or the increase in reactive oxygen species to break bone reconstruction balance. Therefore, regulating the production of inflammatory factors and reactive oxygen species may become a strategy for the treatment of osteoporosis. Solid evidence showed that the overexpression of HO-1 compensates high oxidation levels by increasing intracellular antioxidant levels and reduces inflammation by suppressing pro-inflammatory factors. Some extracts can target HO-1 and ameliorate osteoporosis. However, no systematic report is available on therapies targeting HO-1 to combat osteoporosis. Therefore, this review summarizes the biological characteristics of HO-1, and the relationship between inflammatory response and reactive oxygen species production regulated by HO-1 and osteoporosis. The understanding of the role of HO-1 in osteoporosis may provide ideas for a potential clinical treatment and new drugs targeting HO-1.
Collapse
Affiliation(s)
- Jingmin Che
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong, 518057, China; School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Jiancheng Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Department of Spinal Surgery, People's Hospital of Longhua Shenzhen, Shenzhen, China.
| | - Bin Zhao
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong, 518057, China; School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong, 518057, China; Key Laboratory for Space Bioscience and Biotechnology, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
22
|
Lee TK, Lee JC, Kim DW, Kim B, Sim H, Kim JD, Ahn JH, Park JH, Lee CH, Won MH, Choi SY. Ischemia-Reperfusion under Hyperthermia Increases Heme Oxygenase-1 in Pyramidal Neurons and Astrocytes with Accelerating Neuronal Loss in Gerbil Hippocampus. Int J Mol Sci 2021; 22:ijms22083963. [PMID: 33921375 PMCID: PMC8068892 DOI: 10.3390/ijms22083963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 01/14/2023] Open
Abstract
It has been studied that the damage or death of neurons in the hippocampus is different according to hippocampal subregions, cornu ammonis 1–3 (CA1–3), after transient ischemia in the forebrain, showing that pyramidal neurons located in the subfield CA1 (CA1) are most vulnerable to this ischemia. Hyperthermia is a proven risk factor for brain ischemia and can develop more severe and extensive brain damage related with mortality rate. It is well known that heme oxygenase-1 (HO-1) activity and expression is increased by various stimuli in the brain, including hyperthermia. HO-1 can be either protective or deleterious in the central nervous system, and its roles depend on the expression levels of enzymes. In this study, we investigated the effects of hyperthermia during ischemia on HO-1 expression and neuronal damage/death in the hippocampus to examine the relationship between HO-1 and neuronal damage/death following 5-min transient ischemia in the forebrain using gerbils. Gerbils were assigned to four groups: (1) sham-operated gerbils with normothermia (Normo + sham group); (2) ischemia-operated gerbils with normothermia (Normo + ischemia group); (3) sham-operated gerbils with hyperthermia (39.5 ± 0.2 °C) during ischemia (Hyper + sham group); and (4) ischemia-operated gerbils with hyperthermia during ischemia (Hyper + ischemia group). HO-1 expression levels in CA1–3 of the Hyper + ischemia group were significantly higher than those in the Normo + ischemia group. HO-1 immunoreactivity in the Hyper + ischemia group was significantly increased in pyramidal neurons and astrocytes with time after ischemia, and the immunoreactivity was significantly higher than that in the Normo + ischemia group. In the Normo + Ischemia group, neuronal death was shown in pyramidal neurons located only in CA1 at 5 days after ischemia. However, in the Hyper + ischemia group, pyramidal neuronal death occurred in CA1–3 at 2 days after ischemia. Taken together, our findings showed that brain ischemic insult during hyperthermic condition brings up earlier and severer neuronal damage/death in the hippocampus, showing that HO-1 expression in neurons and astrocytes is different according to brain subregions and temperature condition. Based on these findings, we suggest that hyperthermia in patients with ischemic stroke must be taken into the consideration in the therapy.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Gangwon, Korea;
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung 25457, Gangwon, Korea;
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
| | - Hyejin Sim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
| | - Jong Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon 24341, Gangwon, Korea;
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan 50510, Gyeongnam, Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju 38066, Gyeongbuk, Korea;
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Chungnam, Korea;
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Gangwon, Korea; (J.-C.L.); (B.K.); (H.S.); (J.H.A.)
- Correspondence: (M.-H.W.); (S.Y.C.); Tel.: +82-33-250-8891 (M.-H.W.); +82-33-248-2112 (S.Y.C.); Fax: +82-33-256-1614 (M.-H.W.); +82-33-241-1463 (S.Y.C.)
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Gangwon, Korea;
- Correspondence: (M.-H.W.); (S.Y.C.); Tel.: +82-33-250-8891 (M.-H.W.); +82-33-248-2112 (S.Y.C.); Fax: +82-33-256-1614 (M.-H.W.); +82-33-241-1463 (S.Y.C.)
| |
Collapse
|
23
|
Ishige okamurae Suppresses Trimethyltin-Induced Neurodegeneration and Glutamate-Mediated Excitotoxicity by Regulating MAPKs/Nrf2/HO-1 Antioxidant Pathways. Antioxidants (Basel) 2021; 10:antiox10030440. [PMID: 33809381 PMCID: PMC8001419 DOI: 10.3390/antiox10030440] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Many neurodegenerative diseases have several similar cellular dysregulations. We investigated the inhibitory role of Ishige okamurae, an edible brown alga, on neurodegenerative processes by estimating the effects of Ishige okamurae on excitotoxicity induced by glutamate in vitro and neurodegeneration induced by trimethyltin (TMT) in vivo. This study aimed to describe the molecular mechanisms responsible for the mediating anti-neurodegenerative effects of Ishige okamurae extract (IOE). The oral administration of IOE to TMT-injected mice impeded the TMT-mediated short- and long-term memory impairments investigated by the Morris water maze and Y-maze test. IOE attenuated TMT-mediated cellular apoptosis and the expression of brain-derived neurotrophic factor, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in mice brains. Glutamate-induced apoptosis and the expression of reactive oxygen species, Nrf2, and HO-1 in HT22 cells were also attenuated by IOE. In addition, TMT- and glutamate-induced phosphorylation of mitogen-activated protein kinases (MAPKs) in mouse brain tissues and HT22 cells were attenuated by the treatment of IOE. In HT22 cells, administration of MAPK inhibitors recovered the glutamate induced by the expression of Nrf2, HO-1, and cellular dysregulation to the equal extent to IOE administration. Taken together, these results suggest that IOE could attenuate neurodegenerative processes, such as TMT- and glutamate-mediated neuronal dysregulation, by regulating MAPKs/Nrf-2/HO-1 antioxidant pathways.
Collapse
|
24
|
Gordon JL, Hinsen KJ, Reynolds MM, Smith TA, Tucker HO, Brown MA. Anticancer potential of nitric oxide (NO) in neuroblastoma treatment. RSC Adv 2021; 11:9112-9120. [PMID: 35423416 PMCID: PMC8695301 DOI: 10.1039/d1ra00275a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/18/2021] [Indexed: 11/21/2022] Open
Abstract
The most common extracranial solid tumor in childhood, paediatric neuroblastoma, is frequently diagnosed at advanced stages and identified as high risk. High risk neuroblastoma is aggressive and unpredictable, resulting in poor prognosis and only ∼40% five-year survival rates. Herein, nitric oxide (NO) delivered via the S-nitrosothiol, S-nitrosoglutathione (GSNO), is explored as an anticancer therapeutic in various neuroblastoma lines. After 24 h of treatment with GSNO, cell viability assays, as assessed by resazurin and MTT ((3-4,5-dimethylthiazol-2-yl)-2,5-diphyltetrazolium bromide), consistently identified a moderate, ∼13-29%, decrease in metabolic activity, colony formation assays revealed notably significant reduction of clonogenic activity, and cytotoxicity assays revealed a visibly significant reduction of total number of cells and live cells as well as an increase in number of dead cells in treated cells versus untreated cells. Thrillingly, RNA-sequence analysis provided highly valuable information regarding the differentially expressed genes in treated samples versus control samples as well as insight into the mechanism of action of NO as an anticancer therapeutic. Favorably, the collective results from these analyses exhibited tumoricidal, non-tumour promoting, and discriminatory characteristics, illuminating the feasibility and significance of NO as a cytotoxic adjuvant in neuroblastoma treatment.
Collapse
Affiliation(s)
- Jenna L Gordon
- Department of Chemistry, Colorado State University Fort Collins CO 80521 USA
| | - Kristin J Hinsen
- Department of Biomedical Sciences, Colorado State University Fort Collins CO 80521 USA
| | - Melissa M Reynolds
- Department of Chemistry, School of Biomedical Engineering, Department of Chemical and Biological Engineering, Colorado State University, Campus Delivery 1872 Fort Collins CO 80523 USA
| | - Tyler A Smith
- Department of Neuroscience, University of Texas Austin 2500 Speedway Austin TX 78712 USA
| | - Haley O Tucker
- Department of Molecular Genetics, Institute for Cellular and Molecular Biology, University of Texas Austin Austin TX 78712 USA
| | - Mark A Brown
- Department of Clinical Sciences, Colorado State University Fort Collins CO 80523 USA
| |
Collapse
|
25
|
Zhang L, Zhang X, Wu T, Pan X, Wang Z. Isoflurane reduces septic neuron injury by HO‑1‑mediated abatement of inflammation and apoptosis. Mol Med Rep 2020; 23:155. [PMID: 33355378 PMCID: PMC7789092 DOI: 10.3892/mmr.2020.11794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 11/27/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) frequently occurs in critically ill patients with severe systemic infections. Subanesthetic isoflurane (0.7% ISO) possesses anti-inflammatory, antioxidant and anti-apoptotic properties against a number of human diseases, including brain injury. The activation of heme oxygenase-1 (HO-1) impedes inflammation, oxidation and apoptosis, thus alleviating sepsis-induced brain damage. However, whether 0.7% ISO affords protection against septic neuronal injury involving HO-1 activation is unclear. The present study aimed to investigate the neuroprotective effects of 0.7% ISO and its potential underlying mechanisms in SAE using a mouse model established by cecal ligation and puncture (CLP). The results indicated that the expression and activity of HO-1 in the mouse hippocampus were increased by CLP, and further enhanced by ISO. ISO reduced the death rate, brain water content and blood-brain barrier disruption, but improved the learning and memory functions of CLP-treated mice. ISO significantly decreased the production of pro-inflammatory cytokines and the levels of oxidative indictors in the serum and hippocampus, as well as the number of apoptotic neurons and the expression of pro-apoptotic proteins in the hippocampus. Inversely, anti-inflammatory factors, antioxidative enzymes and anti-apoptotic proteins were markedly increased by ISO administration. However, the neuroprotective effects of ISO were abolished by a HO-1 inhibitor. Overall, these findings suggested that 0.7% ISO alleviated SAE via its anti-inflammatory, antioxidative and anti-apoptotic properties, which involved the activated form of HO-1.
Collapse
Affiliation(s)
- Lina Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xuece Zhang
- Digestive Department, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Ting Wu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xu Pan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zhi Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
26
|
TGF-β1 signaling protects retinal ganglion cells from oxidative stress via modulation of the HO-1/Nrf2 pathway. Chem Biol Interact 2020; 331:109249. [DOI: 10.1016/j.cbi.2020.109249] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/08/2020] [Accepted: 08/27/2020] [Indexed: 01/10/2023]
|
27
|
Iyaswamy A, Krishnamoorthi SK, Liu YW, Song JX, Kammala AK, Sreenivasmurthy SG, Malampati S, Tong BCK, Selvarasu K, Cheung KH, Lu JH, Tan JQ, Huang CY, Durairajan SSK, Li M. Yuan-Hu Zhi Tong Prescription Mitigates Tau Pathology and Alleviates Memory Deficiency in the Preclinical Models of Alzheimer's Disease. Front Pharmacol 2020; 11:584770. [PMID: 33192524 PMCID: PMC7663173 DOI: 10.3389/fphar.2020.584770] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/25/2020] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by memory dysfunction, Aβ plaques together with phosphorylated tau-associated neurofibrillary tangles. Unfortunately, the present existing drugs for AD only offer mild symptomatic cure and have more side effects. As such, developments of effective, nontoxic drugs are immediately required for AD therapy. Present study demonstrates a novel role of Chinese medicine prescription Yuan-Hu Zhi Tong (YZT) in treating AD, and it has substantiated the in vivo effectiveness of YZT in two different transgenic mice models of AD, namely P301S tau and 3XTg-AD mice. Oral treatment of YZT significantly ameliorates motor dysfunction as well as promotes the clearance of aggregated tau in P301S tau mice. YZT improves the cognitive function and reduces the insoluble tau aggregates in 3XTg-AD mice model. Furthermore, YZT decreases the insoluble AT8 positive neuron load in both P301S tau and 3XTg-AD mice. Using microarray and the "Connectivity Map" analysis, we determined the YZT-induced changes in expression of signaling molecules and revealed the potential mechanism of action of YZT. YZT might regulate ubiquitin proteasomal system for the degradation of tau aggregates. The research results show that YZT is a potential drug candidate for the therapy of tau pathogenesis and memory decline in AD.
Collapse
Affiliation(s)
- A Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - S K Krishnamoorthi
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Y W Liu
- Institute of Biopharmaceutical Sciences, National Yang Ming University, Taipei, Taiwan
| | - J X Song
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.,Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - A K Kammala
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - S G Sreenivasmurthy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - S Malampati
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - B C K Tong
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - K Selvarasu
- Division of Mycobiology and Neurodegenerative Disease Research, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - K H Cheung
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - J H Lu
- State Key Lab of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China
| | - J Q Tan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - C Y Huang
- Institute of Biopharmaceutical Sciences, National Yang Ming University, Taipei, Taiwan
| | - S S K Durairajan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.,Division of Mycobiology and Neurodegenerative Disease Research, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - M Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
28
|
Casares L, Unciti-Broceta JD, Prados ME, Caprioglio D, Mattoteia D, Higgins M, Apendino G, Dinkova-Kostova AT, Muñoz E, de la Vega L. Isomeric O-methyl cannabidiolquinones with dual BACH1/NRF2 activity. Redox Biol 2020; 37:101689. [PMID: 32863231 PMCID: PMC7476313 DOI: 10.1016/j.redox.2020.101689] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/31/2020] [Accepted: 08/17/2020] [Indexed: 01/04/2023] Open
Abstract
Oxidative stress and inflammation in the brain are two key hallmarks of neurodegenerative diseases (NDs) such as Alzheimer's, Parkinson's, Huntington's and multiple sclerosis. The axis NRF2-BACH1 has anti-inflammatory and anti-oxidant properties that could be exploited pharmacologically to obtain neuroprotective effects. Activation of NRF2 or inhibition of BACH1 are, individually, promising therapeutic approaches for NDs. Compounds with dual activity as NRF2 activators and BACH1 inhibitors, could therefore potentially provide a more robust antioxidant and anti-inflammatory effects, with an overall better neuroprotective outcome. The phytocannabinoid cannabidiol (CBD) inhibits BACH1 but lacks significant NRF2 activating properties. Based on this scaffold, we have developed a novel CBD derivative that is highly effective at both inhibiting BACH1 and activating NRF2. This new CBD derivative provides neuroprotection in cell models of relevance to Huntington's disease, setting the basis for further developments in vivo.
Collapse
Affiliation(s)
- Laura Casares
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, UK
| | | | | | - Diego Caprioglio
- Dipartimento di Scienze Del Farmaco, Università Del Piemonte Orientale, Novara, Italy
| | - Daiana Mattoteia
- Dipartimento di Scienze Del Farmaco, Università Del Piemonte Orientale, Novara, Italy
| | - Maureen Higgins
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, UK
| | - Giovanni Apendino
- Dipartimento di Scienze Del Farmaco, Università Del Piemonte Orientale, Novara, Italy
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, UK
| | - Eduardo Muñoz
- Instituto Maimónides de Investigación Biomédica de Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Spain; Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Laureano de la Vega
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, UK.
| |
Collapse
|
29
|
Li X, Liu W, Li R, Guo S, Fan H, Wei B, Zhang X, He X, Duan C. TSG-6 Attenuates Oxidative Stress-Induced Early Brain Injury in Subarachnoid Hemorrhage Partly by the HO-1 and Nox2 Pathways. J Stroke Cerebrovasc Dis 2020; 29:104986. [PMID: 32992175 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104986] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/11/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Early brain injury (EBI) refers to acute brain injury during the first 72 h after subarachnoid hemorrhage (SAH), which is one of the major causes of poor prognosis after SAH. Here, we investigated the effect and the related mechanism of TSG-6 on EBI after SAH. MATERIALS AND METHODS The Sprague-Dawley rat model of SAH was developed by the endovascular perforation method. TSG-6 (5μg) was administered by an intraventricular injection within 1.5 h after SAH. The effects of TSG-6 on EBI were assessed by neurological score, brain water content (BWC) and TUNEL staining. Immunofluorescence staining was used to assay NF-κB/p-NF-κB expression in microglia. Protein expression levels of heme oxygenase-1 (HO-1), NADPH oxidase 2 (Nox2), Bcl-2, Bax, and cleaved-caspase-3 were measured to investigate the potential mechanism. The enzyme activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) and the level of reactive oxygen species (ROS) were analyzed using commercially available kits. RESULTS The results showed that TSG-6 treatment alleviated the neurobehavioral dysfunction and reduced BWC and the number of TUNEL-positive neurons in EBI after SAH. TSG-6 decreased the ROS level and enhanced the enzyme activity of SOD and GSH-Px after SAH. Furthermore TSG-6 inhibited the NF-κB activation, increased the protein expression levels of HO-1 and Bcl-2 and decreased the expression levels of Nox2, Bax, and cleaved-caspase-3. The administration of TSG-6 siRNA abolished the protective effects of TSG-6 on EBI after SAH. CONCLUSION We found that TSG-6 attenuated oxidative stress and apoptosis in EBI after SAH partly by inhibiting NF-κB and activating HO-1 pathway in brain tissue.
Collapse
Affiliation(s)
- Xifeng Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Wenchao Liu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Ran Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Shenquan Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Haiyan Fan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Boyang Wei
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Xin Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Xuying He
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Chuanzhi Duan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China.
| |
Collapse
|
30
|
Zhu L, Yi X, Ma C, Luo C, Kong L, Lin X, Gao X, Yuan Z, Wen L, Li R, Wu J, Yi J. Betulinic Acid Attenuates Oxidative Stress in the Thymus Induced by Acute Exposure to T-2 Toxin via Regulation of the MAPK/Nrf2 Signaling Pathway. Toxins (Basel) 2020; 12:toxins12090540. [PMID: 32842569 PMCID: PMC7551141 DOI: 10.3390/toxins12090540] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
T-2 toxin, the most toxic of the trichothecenes, is widely found in grains and feeds, and its intake poses serious risks to the health of humans and animals. An important cytotoxicity mechanism of T-2 toxin is the production of excess free radicals, which in turn leads to oxidative stress. Betulinic acid (BA) has many biological activities, including antioxidant activity, which is a plant-derived pentacyclic triterpenoid. The protective effects and mechanisms of BA in blocking oxidative stress caused by acute exposure to T-2 toxin in the thymus of mice was studied. BA pretreatment reduced ROS production, decreased the MDA content, and increased the content of IgG in serum and the levels of SOD and GSH in the thymus. BA pretreatment also reduced the degree of congestion observed in histopathological tissue sections of the thymus induced by T-2 toxin. Besides, BA downregulated the phosphorylation of the p38, JNK, and ERK proteins, while it upregulated the expression of the Nrf2 and HO-1 proteins in thymus tissues. The results indicated that BA could protect the thymus against the oxidative damage challenged by T-2 toxin by activating Nrf2 and suppressing the MAPK signaling pathway.
Collapse
Affiliation(s)
- Lijuan Zhu
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
| | - Xianglian Yi
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
| | - Chaoyang Ma
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
| | - Chenxi Luo
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
| | - Li Kong
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
| | - Xing Lin
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
| | - Xinyu Gao
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
| | - Zhihang Yuan
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
| | - Lixin Wen
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Rongfang Li
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Jing Wu
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
- Correspondence: (J.W.); (J.Y.)
| | - Jine Yi
- Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (L.Z.); (X.Y.); (C.M.); (C.L.); (L.K.); (X.L.); (X.G.); (Z.Y.); (L.W.); (R.L.)
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Changsha 410128, China
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
- Correspondence: (J.W.); (J.Y.)
| |
Collapse
|
31
|
Role of CGRP in Neuroimmune Interaction via NF-κB Signaling Genes in Glial Cells of Trigeminal Ganglia. Int J Mol Sci 2020; 21:ijms21176005. [PMID: 32825453 PMCID: PMC7503816 DOI: 10.3390/ijms21176005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022] Open
Abstract
Activation of the trigeminal system causes the release of various neuropeptides, cytokines, and other immune mediators. Calcitonin gene-related peptide (CGRP), which is a potent algogenic mediator, is expressed in the peripheral sensory neurons of trigeminal ganglion (TG). It affects the inflammatory responses and pain sensitivity by modulating the activity of glial cells. The primary aim of this study was to use array analysis to investigate the effect of CGRP on the glial cells of TG in regulating nuclear factor kappa B (NF-κB) signaling genes and to further check if CGRP in the TG can affect neuron-glia activation in the spinal trigeminal nucleus caudalis. The glial cells of TG were stimulated with CGRP or Minocycline (Min) + CGRP. The effect on various genes involved in NF-κB signaling pathway was analyzed compared to no treatment control condition using a PCR array analysis. CGRP, Min + CGRP or saline was directly injected inside the TG and the effect on gene expression of Egr1, Myd88 and Akt1 and protein expression of cleaved Caspase3 (cleav Casp3) in the TG, and c-Fos and glial fibrillary acidic protein (GFAP) in the spinal section containing trigeminal nucleus caudalis was analyzed. Results showed that CGRP stimulation resulted in the modulation of several genes involved in the interleukin 1 signaling pathway and some genes of the tumor necrosis factor pathway. Minocycline pre-treatment resulted in the modulation of several genes in the glial cells, including anti-inflammatory genes, and neuronal activation markers. A mild increase in cleav Casp3 expression in TG and c-Fos and GFAP in the spinal trigeminal nucleus of CGRP injected animals was observed. These data provide evidence that glial cells can participate in neuroimmune interaction due to CGRP in the TG via NF-κB signaling pathway.
Collapse
|
32
|
Kim M, Kim H, Kim D, Kim D, Huh Y, Park C, Chung HJ, Jung J, Jeong NY. Heme Oxygenase 1 in Schwann Cells Regulates Peripheral Nerve Degeneration Against Oxidative Stress. ASN Neuro 2020; 11:1759091419838949. [PMID: 31046408 PMCID: PMC6498775 DOI: 10.1177/1759091419838949] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
During Wallerian degeneration, Schwann cells lose their characteristic of myelinating axons and shift into the state of developmental promyelinating cells. This recharacterized Schwann cell guides newly regrowing axons to their destination and remyelinates reinnervated axons. This Schwann cell dynamics during Wallerian degeneration is associated with oxidative events. Heme oxygenases (HOs) are involved in the oxidative degradation of heme into biliverdin/bilirubin, ferrous iron, and carbon monoxide. Overproduction of ferrous iron by HOs increases reactive oxygen species, which have deleterious effects on living cells. Thus, the key molecule for understanding the exact mechanism of Wallerian degeneration in the peripheral nervous system is likely related to oxidative stress-mediated HOs in Schwann cells. In this study, we demonstrate that demyelinating Schwann cells during Wallerian degeneration highly express HO1, not HO2, and remyelinating Schwann cells during nerve regeneration decrease HO1 activation to levels similar to those in normal myelinating Schwann cells. In addition, HO1 activation during Wallerian degeneration regulates several critical phenotypes of recharacterized repair Schwann cells, such as demyelination, transdedifferentiation, and proliferation. Thus, these results suggest that oxidative stress in Schwann cells after peripheral nerve injury may be regulated by HO1 activation during Wallerian degeneration and oxidative-stress-related HO1 activation in Schwann cells may be helpful to study deeply molecular mechanism of Wallerian degeneration.
Collapse
Affiliation(s)
- Muwoong Kim
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Hyosun Kim
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea.,2 Department of Biomedical Science, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Graduation School, Kyung Hee Univeristy, Seoul, Korea
| | - Dogyeong Kim
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea.,2 Department of Biomedical Science, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Graduation School, Kyung Hee Univeristy, Seoul, Korea
| | - Dokyoung Kim
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea.,2 Department of Biomedical Science, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Graduation School, Kyung Hee Univeristy, Seoul, Korea
| | - Youngbuhm Huh
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea.,2 Department of Biomedical Science, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Graduation School, Kyung Hee Univeristy, Seoul, Korea
| | - Chan Park
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea.,2 Department of Biomedical Science, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Graduation School, Kyung Hee Univeristy, Seoul, Korea
| | - Hyung-Joo Chung
- 3 Department of Anesthesiology and Pain Medicine, College of Medicine, Kosin University, Busan, Korea
| | - Junyang Jung
- 1 Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea.,2 Department of Biomedical Science, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Graduation School, Kyung Hee Univeristy, Seoul, Korea
| | - Na Young Jeong
- 4 Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan, Korea
| |
Collapse
|
33
|
Regenerative Potential of Carbon Monoxide in Adult Neural Circuits of the Central Nervous System. Int J Mol Sci 2020; 21:ijms21072273. [PMID: 32218342 PMCID: PMC7177523 DOI: 10.3390/ijms21072273] [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: 02/24/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 01/04/2023] Open
Abstract
Regeneration of adult neural circuits after an injury is limited in the central nervous system (CNS). Heme oxygenase (HO) is an enzyme that produces HO metabolites, such as carbon monoxide (CO), biliverdin and iron by heme degradation. CO may act as a biological signal transduction effector in CNS regeneration by stimulating neuronal intrinsic and extrinsic mechanisms as well as mitochondrial biogenesis. CO may give directions by which the injured neurovascular system switches into regeneration mode by stimulating endogenous neural stem cells and endothelial cells to produce neurons and vessels capable of replacing injured neurons and vessels in the CNS. The present review discusses the regenerative potential of CO in acute and chronic neuroinflammatory diseases of the CNS, such as stroke, traumatic brain injury, multiple sclerosis and Alzheimer’s disease and the role of signaling pathways and neurotrophic factors. CO-mediated facilitation of cellular communications may boost regeneration, consequently forming functional adult neural circuits in CNS injury.
Collapse
|
34
|
Morita A, Jullienne A, Salehi A, Hamer M, Javadi E, Alsarraj Y, Tang J, Zhang JH, Pearce WJ, Obenaus A. Temporal evolution of heme oxygenase-1 expression in reactive astrocytes and microglia in response to traumatic brain injury. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2020.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
35
|
Brandes MS, Gray NE. NRF2 as a Therapeutic Target in Neurodegenerative Diseases. ASN Neuro 2020; 12:1759091419899782. [PMID: 31964153 PMCID: PMC6977098 DOI: 10.1177/1759091419899782] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Increased reactive oxygen species production and oxidative stress have been implicated in the pathogenesis of numerous neurodegenerative conditions including among others Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, Friedrich’s ataxia, multiple sclerosis, and stroke. The endogenous antioxidant response pathway protects cells from oxidative stress by increasing the expression of cytoprotective enzymes and is regulated by the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2). In addition to regulating the expression of antioxidant genes, NRF2 has also been shown to exert anti-inflammatory effects and modulate both mitochondrial function and biogenesis. This is because mitochondrial dysfunction and neuroinflammation are features of many neurodegenerative diseases as well NRF2 has emerged as a promising therapeutic target. Here, we review evidence for a beneficial role of NRF2 in neurodegenerative conditions and the potential of specific NRF2 activators as therapeutic agents.
Collapse
Affiliation(s)
- Mikah S. Brandes
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
36
|
CGRP Reduces Apoptosis of DRG Cells Induced by High-Glucose Oxidative Stress Injury through PI3K/AKT Induction of Heme Oxygenase-1 and Nrf-2 Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2053149. [PMID: 31885775 PMCID: PMC6899316 DOI: 10.1155/2019/2053149] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 10/22/2019] [Indexed: 01/26/2023]
Abstract
Dorsal root ganglion (DRG) neurons, which are sensitive to oxidative stress due to their anatomical and structural characteristics, play a complex role in the initiation and progression of diabetic bladder neuropathy. We investigated the hypothesis that the antioxidant and antiapoptotic effects of CGRP may be partly related to the expression of Nrf2 and HO-1, via the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, thus reducing apoptosis and oxidative stress responses. This study shows that CGRP activates the PI3K/AKT pathway, thereby inducing increased expression of Nrf2 and HO-1 and resulting in the decrease of reactive oxygen species and malondialdehyde levels and reduced neuronal apoptosis. These effects were suppressed by LY294002, an inhibitor of the PI3K/AKT pathway. Therefore, regulation of Nrf2 and HO-1 expression by the PI3K/AKT pathway plays an important role in the regulation of the antioxidant and antiapoptotic responses in DRG cells in a high-glucose culture model.
Collapse
|
37
|
Lim HS, Kim JS, Moon BC, Ryu SM, Lee J, Park G. Batryticatus Bombyx Protects Dopaminergic Neurons Against MPTP-Induced Neurotoxicity by Inhibiting Oxidative Damage. Antioxidants (Basel) 2019; 8:antiox8120574. [PMID: 31766449 PMCID: PMC6943627 DOI: 10.3390/antiox8120574] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023] Open
Abstract
Oxidative stress plays an important role in the degeneration of dopaminergic neurons in Parkinson’s disease (PD). Altered redox homeostasis in neurons interferes with several biological processes, ultimately leading to neuronal death. Oxidative damage has been identified as one of the principal mechanisms underlying the progression of PD. Several studies highlight the key role of superoxide radicals in inducing neuronal toxicity. Batryticatus Bombyx (BB), the dried larva of Bombyx mori L. infected by Beauveria bassiana (Bals.) Vuill., has been used in traditional medicine for its various pharmacological effects. In the present study, BB showed a beneficial effect on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity by directly targeting dopaminergic neurons. Treatment with BB improved behavioral impairments, protected dopaminergic neurons, and maintained dopamine levels in PD mouse models. Here, we investigated the protective effects of BB on MPTP-induced PD in mice and explored the underlying mechanisms of action, focusing on oxidative signaling. In MPTP-induced PD, BB promoted recovery from impaired movement, prevented dopamine depletion, and protected against dopaminergic neuronal degradation in the substantia nigra pars compacta (SNpc) or the striatum (ST). Moreover, BB upregulated mediators of antioxidative response such as superoxidase dismutase (SOD), catalase (CAT), glutathione (GSH), Heme oxygenase 1 (HO-1), and NAD(P)H (nicotinamide adenine dinucleotide phosphate) dehydrogenase (NQO1). Thus, treatment with BB reduced the oxidative stress, improved behavioral impairments, and protected against dopamine depletion in MPTP-induced toxicity.
Collapse
|
38
|
Lee H, Ko W, Chowdhury A, Li B, Kim SC, Oh H, Kim YC, Woo ER, Baek NI, Lee DS. Brassicaphenanthrene A from Brassica rapa protects HT22 neuronal cells through the regulation of Nrf2‑mediated heme oxygenase‑1 expression. Mol Med Rep 2019; 21:493-500. [PMID: 31746357 DOI: 10.3892/mmr.2019.10824] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/09/2019] [Indexed: 11/05/2022] Open
Abstract
Brain cell damage that results from oxidative toxicity contributes to neuronal degeneration. The transcription factor nuclear factor‑E2‑related factor 2 (Nrf2) regulates the expression of heme oxygenase (HO)‑1 and glutathione (GSH), and serves a key role in the pathogenesis of neurological diseases. Brassica rapa is a turnip that is unique to Ganghwa County, and is used mainly for making kimchi, a traditional Korean food. In the current study, brassicaphenanthrene A (BrPA) from B. rapa was demonstrated to exhibit protective effects against neurotoxicity induced by glutamate via Nrf2‑mediated HO‑1 expression. Similarly, BrPA increased the expression of cellular glutathione and glutamine‑cysteine ligase genes. Furthermore, BrPA caused the nuclear translocation of Nrf2 and increased antioxidant response element (ARE) promoter activity. Nrf2 also mediated HO‑1 induction by BrPA through the PI3K/Akt and JNK regulatory pathways. The results of the present study indicated the neuroprotective effect of BrPA, a natural food component from B. rapa.
Collapse
Affiliation(s)
- Hwan Lee
- College of Pharmacy, Chosun University, Dong‑gu, Gwangju 61452, Republic of Korea
| | - Wonmin Ko
- College of Pharmacy, Chosun University, Dong‑gu, Gwangju 61452, Republic of Korea
| | | | - Bin Li
- Department of Pharmacy, Qingdao University of Science and Technology, Qingdao, Shandong 266042, P.R. China
| | - Sam Cheol Kim
- Department of Family Practice and Community Medicine, Chosun University College of Medicine, Dong‑gu, Gwangju 61452, Republic of Korea
| | - Hyuncheol Oh
- College of Pharmacy, Wonkwang University, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Youn-Chul Kim
- College of Pharmacy, Wonkwang University, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Eun-Rhan Woo
- College of Pharmacy, Chosun University, Dong‑gu, Gwangju 61452, Republic of Korea
| | - Nam-In Baek
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung‑Hee University, Yongin, Gyeonggi-do 17104, Republic of Korea
| | - Dong-Sung Lee
- College of Pharmacy, Chosun University, Dong‑gu, Gwangju 61452, Republic of Korea
| |
Collapse
|
39
|
The Nrf2/HO-1 Axis as Targets for Flavanones: Neuroprotection by Pinocembrin, Naringenin, and Eriodictyol. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4724920. [PMID: 31814878 PMCID: PMC6878820 DOI: 10.1155/2019/4724920] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/16/2022]
Abstract
Flavanones are a group of flavonoids that derive from their immediate chalcone precursors through the action of chalcone isomerase enzymes. The Aromatic A and B rings, C4-keto group, and the 15-carbon flavonoid skeleton are all evident in flavanones, but a notable absence of C2-C3 double bond and a lack of oxygenation at C-3 position of the C-ring makes them distinctively different from other groups such as flavonols (e.g., quercetin). On the basis of oxygenation level in the B ring, flavanones can vary from each other as exemplified by pinocembrin (no oxygenation), naringenin (4′-hydroxyl), or eriodictyol (3′,4′-dihydroxyl substitution). These groups are generally weaker free radical scavengers as compared to quercetin and derivatives though eriodictyol has a better free radical scavenging profile within the group due to the presence of the catechol functional moiety. In this communication, their antioxidant potential through the induction of antioxidant defenses is scrutinized. These compounds as exemplified by pinocembrin could induce the nuclear factor erythroid 2-related factor 2- (Nrf2-) heme oxygenase-1 (HO-1) axis leading to amelioration of oxidative stress in cellular and animal models. Their neuroprotective effect through such mechanism is discussed.
Collapse
|
40
|
Shu K, Zhang Y. Protodioscin protects PC12 cells against oxygen and glucose deprivation-induced injury through miR-124/AKT/Nrf2 pathway. Cell Stress Chaperones 2019; 24:1091-1099. [PMID: 31446555 PMCID: PMC6882996 DOI: 10.1007/s12192-019-01031-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/13/2022] Open
Abstract
The purpose of the current study was to demonstrate the neuroprotective effect of protodioscin (Prot) in an in vitro model of ischemia/reperfusion (I/R) and investigate the underlying molecular mechanism. After PC12 cells were exposed to oxygen and glucose deprivation (OGD) reperfusion, PI staining by flow cytometry was used to quantify the rate of apoptosis. The levels of hypoxia-inducible factor 1-alpha (HIF-1α), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA) were determined using commercially available kits. Intracellular reactive oxygen species (ROS) level was detected using the 20,70-dichlorodihy-drofluorescein diacetate (DCFH-DA) fluorescence assay. The expression levels of heat-shock proteins (HSP), PI3K, AKT, Nrf2, and miR-124 were tested by western blot or quantitative PCR. Prot significantly attenuated oxygen-glucose deprivation/reperfusion (OGD/R)-induced apoptotic death. Prot also reduced the oxidative stress as revealed by increasing the activities of SOD and GSH-Px, decreasing the levels of ROS and MDA. Moreover, mechanism investigations suggested that Prot prevented the decrease of HSP70, HSP32 (hemeoxygenase-1, HO-1), and PI3K protein expression, phosphorylation of AKT, and the accumulation of nuclear Nrf2. The level of miR-124 was decreased in PC12 cells, which was also effectively reversed by Prot treatment. Prot protected PC12 cells against OGD/R-induced injury through inhibiting oxidative stress and apoptosis, which could be associated with increasing HSP proteins expression via activating PI3K/AKT/Nrf2 pathway and miR-124 modulation.
Collapse
Affiliation(s)
- Kun Shu
- Department of Medicine, Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Yuelin Zhang
- Department of Neurosurgery, The Third Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta Road., Xi'an City, 710061, Shanxi Province, China.
| |
Collapse
|
41
|
Robicsek SA, Bhattacharya A, Rabai F, Shukla K, Doré S. Blood-Related Toxicity after Traumatic Brain Injury: Potential Targets for Neuroprotection. Mol Neurobiol 2019; 57:159-178. [PMID: 31617072 DOI: 10.1007/s12035-019-01766-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Emergency visits, hospitalizations, and deaths due to traumatic brain injury (TBI) have increased significantly over the past few decades. While the primary early brain trauma is highly deleterious to the brain, the secondary injury post-TBI is postulated to significantly impact mortality. The presence of blood, particularly hemoglobin, and its breakdown products and key binding proteins and receptors modulating their clearance may contribute significantly to toxicity. Heme, hemin, and iron, for example, cause membrane lipid peroxidation, generate reactive oxygen species, and sensitize cells to noxious stimuli resulting in edema, cell death, and increased morbidity and mortality. A wide range of other mechanisms such as the immune system play pivotal roles in mediating secondary injury. Effective scavenging of all of these pro-oxidant and pro-inflammatory metabolites as well as controlling maladaptive immune responses is essential for limiting toxicity and secondary injury. Hemoglobin metabolism is mediated by key molecules such as haptoglobin, heme oxygenase, hemopexin, and ferritin. Genetic variability and dysfunction affecting these pathways (e.g., haptoglobin and heme oxygenase expression) have been implicated in the difference in susceptibility of individual patients to toxicity and may be target pathways for potential therapeutic interventions in TBI. Ongoing collaborative efforts are required to decipher the complexities of blood-related toxicity in TBI with an overarching goal of providing effective treatment options to all patients with TBI.
Collapse
Affiliation(s)
- Steven A Robicsek
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurosurgery, Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Ayon Bhattacharya
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA.,Department of Pharmacology, KPC Medical College, West Bengal University of Health Sciences, Kolkata, West Bengal, India
| | - Ferenc Rabai
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Krunal Shukla
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurology, Psychiatry, Pharmaceutics and Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
42
|
Thabit S, Handoussa H, Roxo M, Cestari de Azevedo B, S E El Sayed N, Wink M. Styphnolobium japonicum (L.) Schott Fruits Increase Stress Resistance and Exert Antioxidant Properties in Caenorhabditis elegans and Mouse Models. Molecules 2019; 24:E2633. [PMID: 31331055 PMCID: PMC6680879 DOI: 10.3390/molecules24142633] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
Styphnolobium japonicum (L.) Schott is a popular Asian tree widely used in traditional medicine. The current study explored the potential stress resistance and antioxidant activities of its fruits. Phytochemical profiling of the hydroalcoholic fruit extract was done via high performance liquid chromatography-photodiode array-electrospray ionization-mass/mass (HPLC-PDA-ESI-MS/MS). Twenty four phenolic constituents were tentatively identified in the extract. The Caenorhabditis elegans (C. elegans) nematode model in addition to trimethyltin (TMT)-induced neurotoxicity mouse model were used for in vivo evaluation of its antioxidant properties. The ability of the extract to enhance stress resistance was manifested through increasing survival rate by 44.7% and decreasing basal reactive oxygen species (ROS) levels by 72.3% in C. elegans. In addition, the extract increased the levels of the stress response enzyme superoxide dismutase-3 (Sod-3) by 55.5% and decreased the expression of heat shock protein-16.2 (Hsp-16.2) in nematodes, which had been challenged by juglone, by 21%. Using a mouse model, the extract significantly decreased the expression of the oxidative stress marker malondialdehyde (MDA). Furthermore, an elevation in the levels of the antioxidant marker glutathione (GSH), SOD and heme oxygenase-1 (HO-1) enzymes were observed. Our findings imply that Styphnolobium japonicum has the potential to be used in future studies focusing on diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Sara Thabit
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Heba Handoussa
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Mariana Roxo
- Biology Department, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg 69120, Germany
| | - Bruna Cestari de Azevedo
- Biology Department, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg 69120, Germany
- Departmento de Biotecnologia em Plantas Medicinais, Universidade de Ribeirão Preto, 14096-900 Ribeirão Preto, Brazil
| | - Nesrine S E El Sayed
- Pharmacology and Toxicology department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Michael Wink
- Biology Department, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg 69120, Germany.
| |
Collapse
|
43
|
Karasev MM, Stepanenko OV, Rumyantsev KA, Turoverov KK, Verkhusha VV. Near-Infrared Fluorescent Proteins and Their Applications. BIOCHEMISTRY (MOSCOW) 2019; 84:S32-S50. [PMID: 31213194 DOI: 10.1134/s0006297919140037] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
High transparency, low light-scattering, and low autofluorescence of mammalian tissues in the near-infrared (NIR) spectral range (~650-900 nm) open a possibility for in vivo imaging of biological processes at the micro- and macroscales to address basic and applied problems in biology and biomedicine. Recently, probes that absorb and fluoresce in the NIR optical range have been engineered using bacterial phytochromes - natural NIR light-absorbing photoreceptors that regulate metabolism in bacteria. Since the chromophore in all these proteins is biliverdin, a natural product of heme catabolism in mammalian cells, they can be used as genetically encoded fluorescent probes, similarly to GFP-like fluorescent proteins. In this review, we discuss photophysical and biochemical properties of NIR fluorescent proteins, reporters, and biosensors and analyze their characteristics required for expression of these molecules in mammalian cells. Structural features and molecular engineering of NIR fluorescent probes are discussed. Applications of NIR fluorescent proteins and biosensors for studies of molecular processes in cells, as well as for tissue and organ visualization in whole-body imaging in vivo, are described. We specifically focus on the use of NIR fluorescent probes in advanced imaging technologies that combine fluorescence and bioluminescence methods with photoacoustic tomography.
Collapse
Affiliation(s)
- M M Karasev
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia. .,Medicum, University of Helsinki, Helsinki, 00290, Finland
| | - O V Stepanenko
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia.
| | - K A Rumyantsev
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia. .,Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Loginov Moscow Clinical Scientific Center, Moscow, 111123, Russia
| | - K K Turoverov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia. .,Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 195251, Russia
| | - V V Verkhusha
- Medicum, University of Helsinki, Helsinki, 00290, Finland. .,Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
44
|
Habtemariam S. Antioxidant and Anti-inflammatory Mechanisms of Neuroprotection by Ursolic Acid: Addressing Brain Injury, Cerebral Ischemia, Cognition Deficit, Anxiety, and Depression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8512048. [PMID: 31223427 PMCID: PMC6541953 DOI: 10.1155/2019/8512048] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/27/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022]
Abstract
Ursolic acid (UA) is a pentacyclic triterpene which is found in common herbs and medicinal plants that are reputed for a variety of pharmacological effects. Both as an active principle of these plants and as a nutraceutical ingredient, the pharmacology of UA in the CNS and other organs and systems has been extensively reported in recent years. In this communication, the antioxidant and anti-inflammatory axis of UA's pharmacology is appraised for its therapeutic potential in some common CNS disorders. Classic examples include the traumatic brain injury (TBI), cerebral ischemia, cognition deficit, anxiety, and depression. The pharmacological efficacy for UA is demonstrated through the therapeutic principle of one drug → multitargets → one/many disease(s). Both specific enzymes and receptor targets along with diverse pharmacological effects associated with oxidative stress and inflammatory signalling are scrutinised.
Collapse
Affiliation(s)
- Solomon Habtemariam
- Pharmacognosy Research Laboratories & Herbal Analysis Services, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| |
Collapse
|
45
|
Cai M, Zhu Y, Li Z, Josephs-Spaulding J, Zhou Y, Hu Y, Chen H, Liu Y, He W, Zhang J. Profiling the Gene Expression and DNA Methylation in the Mouse Brain after Ischemic Preconditioning. Neuroscience 2019; 406:249-261. [DOI: 10.1016/j.neuroscience.2019.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/09/2019] [Accepted: 03/11/2019] [Indexed: 01/27/2023]
|
46
|
Vasconcelos AR, Dos Santos NB, Scavone C, Munhoz CD. Nrf2/ARE Pathway Modulation by Dietary Energy Regulation in Neurological Disorders. Front Pharmacol 2019; 10:33. [PMID: 30778297 PMCID: PMC6369171 DOI: 10.3389/fphar.2019.00033] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/14/2019] [Indexed: 12/16/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) regulates the expression of an array of enzymes with important detoxifying and antioxidant functions. Current findings support the role of high levels of oxidative stress in the pathogenesis of neurological disorders. Given the central role played by Nrf2 in counteracting oxidative damage, a number of studies have targeted the modulation of this transcription factor in order to confer neuroprotection. Nrf2 activity is tightly regulated by oxidative stress and energy-based stimuli. Thus, many dietary interventions based on energy intake regulation, such as dietary energy restriction (DER) or high-fat diet (HFD), modulate Nrf2 with consequences for a variety of cellular processes that affect brain health. DER, by either restricting calorie intake or meal frequency, activates Nrf2 thereby triggering its protective effects, whilst HFD inhibit this pathway, thereby exacerbating oxidative stress. Consequently, DER protocols can be valuable strategies in the management of central nervous system (CNS) disorders. Herein, we review current knowledge of the role of Nrf2 signaling in neurological diseases, namely Alzheimer’s disease, Parkinson’s disease, multiple sclerosis and cerebral ischemia, as well as the potential of energy intake regulation in the management of Nrf2 signaling.
Collapse
Affiliation(s)
- Andrea Rodrigues Vasconcelos
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Nilton Barreto Dos Santos
- Laboratory of Neuroendocrinopharmacology and Immunomodulation, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Carolina Demarchi Munhoz
- Laboratory of Neuroendocrinopharmacology and Immunomodulation, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
47
|
Low serum bilirubin levels contribute to the presence and progression of distal symmetrical polyneuropathy in Chinese patients with type 2 diabetes. DIABETES & METABOLISM 2019; 45:47-52. [DOI: 10.1016/j.diabet.2018.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 02/10/2018] [Accepted: 02/18/2018] [Indexed: 11/22/2022]
|
48
|
Liu G, Zhou Y, Chen L. Intestinal uptake of barley protein-based nanoparticles for β-carotene delivery. Acta Pharm Sin B 2019; 9:87-96. [PMID: 30766780 PMCID: PMC6362262 DOI: 10.1016/j.apsb.2018.10.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/16/2018] [Accepted: 08/22/2018] [Indexed: 01/10/2023] Open
Abstract
Our previous study introduced a barley protein microparticle for encapsulation of hydrophobic drug/nutraceutical, which could release nanoparticles upon gastric digestion and deliver encapsulated compound to a simulated intestinal environment intact. This work focused on evaluating the potential of liberated nanoparticles to improve the absorption of encapsulated compounds (e.g., β-carotene) using in vitro Caco-2 cell and ex vivo small intestine models. Nanoparticles obtained from gastric digestion of barley protein microparticles had a spherical shape and an average size of 351 nm. Nanoparticles showed low cytotoxicity in Caco-2 cells and their cellular uptake was dependent on time, concentration and temperature. In a Caco-2 cell monolayer model, significantly greater uptake and transport of β-carotene were observed when it was delivered by nanoparticles (15%), compared to free β-carotene suspension (2.6%). In an ex vivo rat jejunum model, nanoparticles showed the capacity to retain in small intestinal tissue. Approximately 2.24 and 6.04 μg nanoparticle were able to permeate through each cm2 intestinal tissue and translocate to the serosal side after 60 and 90 min, respectively. Results from this study demonstrated the absorption improving effect of the barley protein nanoparticles and suggested their potential as vehicles for hydrophobic compounds.
Collapse
|
49
|
The sinister face of heme oxygenase-1 in brain aging and disease. Prog Neurobiol 2019; 172:40-70. [DOI: 10.1016/j.pneurobio.2018.06.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 11/23/2022]
|
50
|
Lee H, Choi YK. Regenerative Effects of Heme Oxygenase Metabolites on Neuroinflammatory Diseases. Int J Mol Sci 2018; 20:ijms20010078. [PMID: 30585210 PMCID: PMC6337166 DOI: 10.3390/ijms20010078] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/13/2022] Open
Abstract
Heme oxygenase (HO) catabolizes heme to produce HO metabolites, such as carbon monoxide (CO) and bilirubin (BR), which have gained recognition as biological signal transduction effectors. The neurovascular unit refers to a highly evolved network among endothelial cells, pericytes, astrocytes, microglia, neurons, and neural stem cells in the central nervous system (CNS). Proper communication and functional circuitry in these diverse cell types is essential for effective CNS homeostasis. Neuroinflammation is associated with the vascular pathogenesis of many CNS disorders. CNS injury elicits responses from activated glia (e.g., astrocytes, oligodendrocytes, and microglia) and from damaged perivascular cells (e.g., pericytes and endothelial cells). Most brain lesions cause extensive proliferation and growth of existing glial cells around the site of injury, leading to reactions causing glial scarring, which may act as a major barrier to neuronal regrowth in the CNS. In addition, damaged perivascular cells lead to the breakdown of the blood-neural barrier, and an increase in immune activation, activated glia, and neuroinflammation. The present review discusses the regenerative role of HO metabolites, such as CO and BR, in various vascular diseases of the CNS such as stroke, traumatic brain injury, diabetic retinopathy, and Alzheimer's disease, and the role of several other signaling molecules.
Collapse
Affiliation(s)
- Huiju Lee
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Yoon Kyung Choi
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|