1
|
Ye X, Li M, Meng Y, Duan S, Zhang S, Zhong K. D-tetramethrin induces cardiac looping failure in zebrafish during embryonic development. Comp Biochem Physiol C Toxicol Pharmacol 2025; 294:110193. [PMID: 40086682 DOI: 10.1016/j.cbpc.2025.110193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/23/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Cardiac looping is a crucial process during embryonic development. Abnormalities or failures in cardiac looping can lead to congenital heart defects, thereby affecting normal physiological function. Environmental pollutant exposure is one of the major causes of cardiac looping failure. D-tetramethrin is a hygienic insecticide widely used in households and public places, that can enter the human body through contact, insect transmission, and the food chain, thereby impacting human health. In this study, zebrafish embryos were exposed to different concentrations of D-tetramethrin to analyze its effects on heart development, and oxidative stress levels within the embryos. Additionally, qPCR was employed to analyze the transcription and the expression levels of genes related to heart development and function. The results showed that (1) D-tetramethrin exposure significantly reduced heart rate and increased the distance between the sinus venosus and the bulbus arteriosus (SV-BA), which suggested that D-tetramethrin induced cardiac looping failure and led to abnormal heart function. (2) D-tetramethrin exposure elevated the levels of reactive oxygen species (ROS) and malondialdehyde (MDA) in zebrafish embryos while decreasing the enzyme activities of key antioxidant stress enzymes, such as catalase (CAT) and superoxide dismutase (SOD). (3) D-tetramethrin exposure resulted in a significant downregulation of the transcription of cardiac looping-related genes (Myh6, Nkx2.5, Tbx2b, Tbx5a, Tnnt2c and Hand2) and heart function-related genes (Gata4, Vmhc and Nppa). Our findings indicate that D-tetramethrin causes the accumulation of ROS, which in turn alters the transcription levels of genes related to cardiac looping, ultimately resulting in cardiac looping failure.
Collapse
Affiliation(s)
- Xinhao Ye
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - Mijia Li
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - YunLong Meng
- School of Medicine, Tongji University, Shanghai 200000, China
| | - Shiyi Duan
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - Sijie Zhang
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - Keyuan Zhong
- Jiangxi Province Key Laboratory of Synthetic Pharmaceutical Chemistry, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China.
| |
Collapse
|
2
|
Sanchez KL, Kim J, White JB, Tolan A, Rajagopal NP, Anderson DW, Shin AN, Shin SD, Currais A, Soriano-Castell D, Maher P, Soriano S. Evidence of Oxytosis/Ferroptosis in Niemann-Pick Disease Type C. Int J Mol Sci 2025; 26:2915. [PMID: 40243519 PMCID: PMC11988824 DOI: 10.3390/ijms26072915] [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: 02/24/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Niemann-Pick Disease Type C (NPC) is a hereditary neurodegenerative disease characterized by selective cell vulnerability, particularly affecting cerebellar anterior Purkinje neurons. These neurons exhibit a distinctive pattern of degeneration due to the loss of NPC1 and/or NPC2 protein function, progressively extending towards posterior cerebellar regions. Our study aimed to explore the early factors influencing this selective vulnerability of anterior Purkinje neurons in NPC. Oxytosis/ferroptosis, a novel form of regulated cell death, has been implicated in neurodegenerative diseases, with its inhibition showing promising therapeutic potential. Our laboratory has previously identified parallels between NPC cellular pathology and ferroptotic markers, including elevated levels of lipid peroxidation and iron, mitochondrial dysfunction, and Ca2+ dyshomeostasis. However, whether oxytosis/ferroptosis underlies NPC cellular pathology remains unexplored. We hypothesize that loss of NPC1 function increases vulnerability to ferroptosis and that anti-ferroptotic compounds will reverse NPC cellular pathology. Through bioinformatic analyses of pre-symptomatic Npc1-/- Purkinje neurons and in vitro studies using primary dermal fibroblasts derived from NPC patients, we provide evidence suggesting that oxytosis/ferroptosis may play a pathogenic role in NPC. These findings highlight the potential of anti-ferroptotic compounds as a promising therapeutic strategy to mitigate neurodegeneration in NPC and potentially other related disorders.
Collapse
Affiliation(s)
- Kayla L. Sanchez
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Jeanyoung Kim
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Jacob B. White
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Andrew Tolan
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Naren P. Rajagopal
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Douglas W. Anderson
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Alexandra N. Shin
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Samuel D. Shin
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Antonio Currais
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - David Soriano-Castell
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Pamela Maher
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Salvador Soriano
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| |
Collapse
|
3
|
Rajan A, Fame RM. Brain development and bioenergetic changes. Neurobiol Dis 2024; 199:106550. [PMID: 38849103 PMCID: PMC11495523 DOI: 10.1016/j.nbd.2024.106550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024] Open
Abstract
Bioenergetics describe the biochemical processes responsible for energy supply in organisms. When these changes become dysregulated in brain development, multiple neurodevelopmental diseases can occur, implicating bioenergetics as key regulators of neural development. Historically, the discovery of disease processes affecting individual stages of brain development has revealed critical roles that bioenergetics play in generating the nervous system. Bioenergetic-dependent neurodevelopmental disorders include neural tube closure defects, microcephaly, intellectual disability, autism spectrum disorders, epilepsy, mTORopathies, and oncogenic processes. Developmental timing and cell-type specificity of these changes determine the long-term effects of bioenergetic disease mechanisms on brain form and function. Here, we discuss key metabolic regulators of neural progenitor specification, neuronal differentiation (neurogenesis), and gliogenesis. In general, transitions between glycolysis and oxidative phosphorylation are regulated in early brain development and in oncogenesis, and reactive oxygen species (ROS) and mitochondrial maturity play key roles later in differentiation. We also discuss how bioenergetics interface with the developmental regulation of other key neural elements, including the cerebrospinal fluid brain environment. While questions remain about the interplay between bioenergetics and brain development, this review integrates the current state of known key intersections between these processes in health and disease.
Collapse
Affiliation(s)
- Arjun Rajan
- Developmental Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Ryann M Fame
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
Mora-Zenil J, Morán J. ROS produced by NOX promote the neurite growth in a PI3K/Akt independent manner. J Neurosci Res 2024; 102:e25259. [PMID: 37840360 DOI: 10.1002/jnr.25259] [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: 11/09/2022] [Revised: 05/17/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
Reactive oxygen species (ROS) function as signaling molecules in several physiologic and pathologic processes. In central nervous system, ROS are critical for differentiation, migration, polarization, and neurite growth. These actions are mediated by reversible oxidation of target proteins. On the other hand, PI3K/Akt signaling pathway is susceptible to be modulated by ROS and it has been implicated in neurite growth. In this study, we evaluated the participation of ROS in the neurite growth of cultured rat cerebellar granule neurons (CGN), as well as the possible regulation of the PI3K/Akt pathway by ROS during neurite outgrowth. For this purpose, CGN were treated with cellular or mitochondrial antioxidants, or an NOX inhibitor and neurite growth was evaluated. Moreover, to assess the participation Akt in this process, the p-Akt levels were measured in CGN treated with antioxidants or a NOX inhibitor. The effect of antioxidants on the neurite growth in the presence of a PI3K inhibitor was also measured. We found that cellular antioxidants and the NOX inhibitor decreased the neurite growth, but not the mitochondrial antioxidant. Interestingly, the antioxidants increased the p-Akt levels; however, the effect of antioxidants on neurite growth was no dependent on the Akt activity since the inhibitor of PI3K did not modify the antioxidant action on neurite growth. Our results show that the PI3K/Akt pathway participates in neurite growth and that ROS produced by NOX could function as signals in this process; however, this action is not mediated by a redox regulation of Akt activity.
Collapse
Affiliation(s)
- Janeth Mora-Zenil
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
5
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [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: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
6
|
Redox-dependent Igfbp2 signaling controls Brca1 DNA damage response to govern neural stem cell fate. Nat Commun 2023; 14:444. [PMID: 36707536 PMCID: PMC9883463 DOI: 10.1038/s41467-023-36174-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/17/2023] [Indexed: 01/29/2023] Open
Abstract
Neural stem cell (NSC) maintenance and functions are regulated by reactive oxygen species (ROS). However, the mechanisms by which ROS control NSC behavior remain unclear. Here we report that ROS-dependent Igfbp2 signaling controls DNA repair pathways which balance NSC self-renewal and lineage commitment. Ncf1 or Igfbp2 deficiency constrains NSCs to a self-renewing state and prevents neurosphere formation. Ncf1-dependent oxidation of Igfbp2 promotes neurogenesis by NSCs in vitro and in vivo while repressing Brca1 DNA damage response genes and inducing DNA double-strand breaks (DDSBs). By contrast, Ncf1-/- and Igfbp2-/- NSCs favor the formation of oligodendrocytes in vitro and in vivo. Notably, transient repression of Brca1 DNA repair pathway genes induces DDSBs and is sufficient to rescue the ability of Ncf1-/- and Igfbp2-/- NSCs to lineage-commit to form neurospheres and neurons. NSC lineage commitment is dependent on the oxidizable cysteine-43 residue of Igfbp2. Our study highlights the role of DNA damage/repair in orchestrating NSC fate decisions downstream of redox-regulated Igfbp2.
Collapse
|
7
|
Hou Y, Wei W, Li G, Sang N. Prenatal PM 2.5 exposure contributes to neuronal tau lesion in male offspring mice through mitochondrial dysfunction-mediated insulin resistance. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 246:114151. [PMID: 36228359 DOI: 10.1016/j.ecoenv.2022.114151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 06/16/2023]
Abstract
The epidemiological evidence has linked prenatal exposure to fine particulate matter (PM2.5) pollution with neurological diseases in offspring. However, the biological process and toxicological mechanisms remain unclear. Tau protein is a neuronal microtubule-associated protein expressed in fetal brain and plays a critical role in mediating neuronal development. Aberrant expression of tau is associated with adverse neurodevelopmental outcomes. To study whether prenatal exposure to PM2.5 pollution induce tau lesion in mice offspring and elucidate the underlying pathogenic mechanism, we exposed pregnant mice to PM2.5 (3 mg/kg b.w.) by oropharyngeal aspiration every other day. The results indicate that prenatal PM2.5 exposure induced hyperphosphorylation of tau in the cortex of postnatal male offspring, which was accompanied by insulin resistance through the IRS-1/PI3K/AKT signaling pathway. Importantly, we further found that prenatal PM2.5 exposure induced mitochondrial dysfunction by disrupting mitochondrial ultrastructure and decreasing the expression of rate-limiting enzymes (CS, IDH2 and FH) in the Krebs cycle and the subunits of mitochondrial complex IV and V (CO1, CO4, ATP6, and ATP8) during postnatal neurodevelopment. The findings suggest that prenatal PM2.5 exposure could induce tauopathy-like changes in male offspring, in which mitochondrial dysfunction-induced insulin resistance might play an important role.
Collapse
Affiliation(s)
- Yanwen Hou
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Wei Wei
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
8
|
Fame RM, Lehtinen MK. Mitochondria in Early Forebrain Development: From Neurulation to Mid-Corticogenesis. Front Cell Dev Biol 2021; 9:780207. [PMID: 34888312 PMCID: PMC8650308 DOI: 10.3389/fcell.2021.780207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/10/2021] [Indexed: 01/07/2023] Open
Abstract
Function of the mature central nervous system (CNS) requires a substantial proportion of the body’s energy consumption. During development, the CNS anlage must maintain its structure and perform stage-specific functions as it proceeds through discrete developmental stages. While key extrinsic signals and internal transcriptional controls over these processes are well appreciated, metabolic and mitochondrial states are also critical to appropriate forebrain development. Specifically, metabolic state, mitochondrial function, and mitochondrial dynamics/localization play critical roles in neurulation and CNS progenitor specification, progenitor proliferation and survival, neurogenesis, neural migration, and neurite outgrowth and synaptogenesis. With the goal of integrating neurodevelopmental biologists and mitochondrial specialists, this review synthesizes data from disparate models and processes to compile and highlight key roles of mitochondria in the early development of the CNS with specific focus on forebrain development and corticogenesis.
Collapse
Affiliation(s)
- Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, MA, United States
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
9
|
Mokhtar HEL, Hulail MAE, Mahmoud SM, Yousef DM. Impact of cisplatin administration on cerebellar cortical structure and locomotor activity of infantile and juvenile albino rats: the role of oxidative stress. Anat Sci Int 2021; 97:30-47. [PMID: 34386931 DOI: 10.1007/s12565-021-00624-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/01/2021] [Indexed: 11/30/2022]
Abstract
The central neurotoxicity of cisplatin (CisPt) has always raised questions especially during development, but few studies are available. Hence, this work was designed to assess the CisPt's impacts on the postnatal rat cerebellum via evaluation of locomotor activity, histological and immunohistochemical studies, and to focus on cerebellar oxidative stress-related alterations. Eighty newborn pups were divided into 2 equal experimental groups: the control group was kept without any treatment and CisPt-treated group received a single subcutaneous injection of CisPt (5 μg /g b.w.) in their nape at PD10. Ten rats at PD11, PD17, and PD30 ages were weighed, then deeply anesthetized and sacrificed. For locomotor assessment, 20 pups were divided equally into control and CisPt-treated groups and tested at PD11-13, PD15-17, and PD28-30 ages. CisPt-treated rats suffered from decreased motor activity and showed decreased body and cerebellar weights, reduced levels of enzymatic antioxidants (SOD and CAT), and non-enzymatic antioxidant defense (GSH), and increase of lipid peroxidation marker (MDA). Histopathologically, CisPt sowed deleterious changes within cerebellar cortical layers in the form of vacuolations, decreased thickness, and hemorrhage (in PD17), while Purkinje cells exhibited profound degenerative changes in the form of swelling, disrupted arrangement, distortion, and nuclear shrinkage. In CisPt-treated rats, GFAP demonstrated upregulated, hypertrophied, and branched Bergmann glial fibers and reactive astrogliosis. Immuno-localization of Ki-67-positive cells revealed defective migration associated with decreased proliferation in early ages in addition to glial proliferation in PD30. In conclusion, CisPt causes oxidative stress-related deleterious effects on structure of developing cerebellar cortex and affects locomotor activity.
Collapse
Affiliation(s)
- Hanan E L Mokhtar
- Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohey A E Hulail
- Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samar Mortada Mahmoud
- Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Doaa Mohammed Yousef
- Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
10
|
Maraldi T, Angeloni C, Prata C, Hrelia S. NADPH Oxidases: Redox Regulators of Stem Cell Fate and Function. Antioxidants (Basel) 2021; 10:973. [PMID: 34204425 PMCID: PMC8234808 DOI: 10.3390/antiox10060973] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
One of the major sources of reactive oxygen species (ROS) generated within stem cells is the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes (NOXs), which are critical determinants of the redox state beside antioxidant defense mechanisms. This balance is involved in another one that regulates stem cell fate: indeed, self-renewal, proliferation, and differentiation are decisive steps for stem cells during embryo development, adult tissue renovation, and cell therapy application. Ex vivo culture-expanded stem cells are being investigated for tissue repair and immune modulation, but events such as aging, senescence, and oxidative stress reduce their ex vivo proliferation, which is crucial for their clinical applications. Here, we review the role of NOX-derived ROS in stem cell biology and functions, focusing on positive and negative effects triggered by the activity of different NOX isoforms. We report recent findings on downstream molecular targets of NOX-ROS signaling that can modulate stem cell homeostasis and lineage commitment and discuss the implications in ex vivo expansion and in vivo engraftment, function, and longevity. This review highlights the role of NOX as a pivotal regulator of several stem cell populations, and we conclude that these aspects have important implications in the clinical utility of stem cells, but further studies on the effects of pharmacological modulation of NOX in human stem cells are imperative.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
| | - Cristina Angeloni
- School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum—University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy;
| |
Collapse
|
11
|
Gindri dos Santos B, Peres Klein C, Scortegagna Crestani M, Moura Maurmann R, Mateus Hözer R, dos Santos Rodrigues K, Maciel August P, Matté C. Naringin Supplementation during Pregnancy Induces Sex and Region-Specific Alterations in the Offspring's Brain Redox Status. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18094805. [PMID: 33946307 PMCID: PMC8124438 DOI: 10.3390/ijerph18094805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 11/26/2022]
Abstract
Research has shown the beneficial effects of naringin supplementation to adult rodents, which can ameliorate oxidative stress in disease models. However, evidence has demonstrated that polyphenol supplementation induced detrimental effects when consumed during sensitive periods of development, such as pregnancy. Therefore, we investigated the effect of maternal naringin supplementation during pregnancy on the offspring’s cerebral redox status. Pregnant Wistar rats were divided into control and naringin groups and supplemented from gestational day 15 to gestational day 21. On postnatal days 1, 7, and 21, offspring were euthanized, and the prefrontal cortex, hippocampus, striatum, and cerebellum dissected. On postnatal day 1, maternal naringin supplementation positively modulated the pups’ brain redox status. On postnatal day 7, a pro-oxidative milieu was observed in the offspring’s striatum and cerebellum in a sex-dependent manner, even though the prefrontal cortex and hippocampus were not negatively affected. Besides, the alterations observed on postnatal day 7 did not persist up to weaning. Our findings demonstrated that the effect induced by naringin supplementation in the brain redox status differed according to the period of development in which naringin was consumed since the beneficial effects usually found in the adult rodents became detrimental when the supplementation was applied during pregnancy.
Collapse
Affiliation(s)
- Bernardo Gindri dos Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (C.P.K.); (R.M.H.); (K.d.S.R.); (P.M.A.); (C.M.)
- Correspondence:
| | - Caroline Peres Klein
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (C.P.K.); (R.M.H.); (K.d.S.R.); (P.M.A.); (C.M.)
| | - Mariana Scortegagna Crestani
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (M.S.C.); (R.M.M.)
| | - Rafael Moura Maurmann
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (M.S.C.); (R.M.M.)
| | - Régis Mateus Hözer
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (C.P.K.); (R.M.H.); (K.d.S.R.); (P.M.A.); (C.M.)
| | - Karoline dos Santos Rodrigues
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (C.P.K.); (R.M.H.); (K.d.S.R.); (P.M.A.); (C.M.)
| | - Pauline Maciel August
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (C.P.K.); (R.M.H.); (K.d.S.R.); (P.M.A.); (C.M.)
| | - Cristiane Matté
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (C.P.K.); (R.M.H.); (K.d.S.R.); (P.M.A.); (C.M.)
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-000, Brazil; (M.S.C.); (R.M.M.)
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90050-170, Brazil
| |
Collapse
|
12
|
Terzi A, Alam SMS, Suter DM. ROS Live Cell Imaging During Neuronal Development. J Vis Exp 2021. [PMID: 33645566 DOI: 10.3791/62165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Reactive oxygen species (ROS) are well-established signaling molecules, which are important in normal development, homeostasis, and physiology. Among the different ROS, hydrogen peroxide (H2O2) is best characterized with respect to roles in cellular signaling. H2O2 has been implicated during the development in several species. For example, a transient increase in H2O2 has been detected in zebrafish embryos during the first days following fertilization. Furthermore, depleting an important cellular H2O2 source, NADPH oxidase (NOX), impairs nervous system development such as the differentiation, axonal growth, and guidance of retinal ganglion cells (RGCs) both in vivo and in vitro. Here, we describe a method for imaging intracellular H2O2 levels in cultured zebrafish neurons and whole larvae during development using the genetically encoded H2O2-specific biosensor, roGFP2-Orp1. This probe can be transiently or stably expressed in zebrafish larvae. Furthermore, the ratiometric readout diminishes the probability of detecting artifacts due to differential gene expression or volume effects. First, we demonstrate how to isolate and culture RGCs derived from zebrafish embryos that transiently express roGFP2-Orp1. Then, we use whole larvae to monitor H2O2 levels at the tissue level. The sensor has been validated by the addition of H2O2. Additionally, this methodology could be used to measure H2O2 levels in specific cell types and tissues by generating transgenic animals with tissue-specific biosensor expression. As zebrafish facilitate genetic and developmental manipulations, the approach demonstrated here could serve as a pipeline to test the role of H2O2 during neuronal and general embryonic development in vertebrates.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Biological Sciences, Purdue University; Purdue Institute for Integrative Neuroscience, Purdue University
| | - S M Sabbir Alam
- Department of Biological Sciences, Purdue University; Purdue Institute for Integrative Neuroscience, Purdue University
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University; Purdue Institute for Integrative Neuroscience, Purdue University; Bindley Bioscience Center, Purdue University;
| |
Collapse
|
13
|
Zhang X, Ibi M, Haga R, Iwata K, Matsumoto M, Asaoka N, Liu J, Katsuyama M, Yabe-Nishimura C. NOX1/NADPH oxidase affects the development of autism-like behaviors in a maternal immune activation model. Biochem Biophys Res Commun 2021; 534:59-66. [PMID: 33310189 DOI: 10.1016/j.bbrc.2020.11.070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 12/26/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder caused by genetic and environmental factors. Among the environmental factors, maternal infection is known as one of the principal risk factors for ASD. On the other hand, postmortem studies suggested the relationship of oxidative stress with ASD etiology. However, the role of oxidative stress in the development of ASD remains unclear. Here, we report the involvement of NOX1/NADPH oxidase, an enzyme generating reactive oxygen species (ROS), in behavioral and anatomical abnormalities in a maternal immune activation (MIA) model. In the MIA model of gestational polyinosinic-polycytidylic acid (poly(I:C)) exposure, increased serum levels of IL-6 were observed in both wild-type (WT) and Nox1-deficient mice (Nox1KO). Following the comparable induction of MIA in the two genotypes, impairment of social preference and defects in motor coordination were observed in WT offspring but not in offspring deficient in Nox1. MIA up-regulated NOX1 mRNA in the cerebral cortex and cerebellum of the fetus but not in the adult offspring. Although the development of cortical neurons was unaffected by MIA in either genotype, the dropout of Purkinje cells in lobule VII of MIA-affected offspring was significantly ameliorated in Nox1KO. Taken together, these results suggested that NOX1/NADPH oxidase plays an essential role in some behavioral phenotypes observed in ASD, possibly by promoting the loss of Purkinje cells in the cerebellum.
Collapse
Affiliation(s)
- Xueqing Zhang
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masakazu Ibi
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Ryu Haga
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kazumi Iwata
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Misaki Matsumoto
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Nozomi Asaoka
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Junjie Liu
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masato Katsuyama
- Radioisotope Center, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Chihiro Yabe-Nishimura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.
| |
Collapse
|
14
|
Terzi A, Roeder H, Weaver CJ, Suter DM. Neuronal NADPH oxidase 2 regulates growth cone guidance downstream of slit2/robo2. Dev Neurobiol 2020; 81:3-21. [PMID: 33191581 DOI: 10.1002/dneu.22791] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/10/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
NADPH oxidases (Nox) are membrane-bound multi-subunit protein complexes producing reactive oxygen species (ROS) that regulate many cellular processes. Emerging evidence suggests that Nox-derived ROS also control neuronal development and axonal outgrowth. However, whether Nox act downstream of receptors for axonal growth and guidance cues is presently unknown. To answer this question, we cultured retinal ganglion cells (RGCs) derived from zebrafish embryos and exposed these neurons to netrin-1, slit2, and brain-derived neurotrophic factor (BDNF). To test the role of Nox in cue-mediated growth and guidance, we either pharmacologically inhibited Nox or investigated neurons from mutant fish that are deficient in Nox2. We found that slit2-mediated growth cone collapse, and axonal retraction were eliminated by Nox inhibition. Though we did not see an effect of either BDNF or netrin-1 on growth rates, growth in the presence of netrin-1 was reduced by Nox inhibition. Furthermore, attractive and repulsive growth cone turning in response to gradients of BDNF, netrin-1, and slit2, respectively, were eliminated when Nox was inhibited in vitro. ROS biosensor imaging showed that slit2 treatment increased growth cone hydrogen peroxide levels via mechanisms involving Nox2 activation. We also investigated the possible relationship between Nox2 and slit2/Robo2 signaling in vivo. astray/nox2 double heterozygote larvae exhibited decreased area of tectal innervation as compared to individual heterozygotes, suggesting both Nox2 and Robo2 are required for establishment of retinotectal connections. Our results provide evidence that Nox2 acts downstream of slit2/Robo2 by mediating growth and guidance of developing zebrafish RGC neurons.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Haley Roeder
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Cory J Weaver
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA.,Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA.,Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
15
|
Terzi A, Suter DM. The role of NADPH oxidases in neuronal development. Free Radic Biol Med 2020; 154:33-47. [PMID: 32370993 DOI: 10.1016/j.freeradbiomed.2020.04.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) are critical for maintaining cellular homeostasis and function when produced in physiological ranges. Important sources of cellular ROS include NADPH oxidases (Nox), which are evolutionary conserved multi-subunit transmembrane proteins. Nox-mediated ROS regulate variety of biological processes including hormone synthesis, calcium signaling, cell migration, and immunity. ROS participate in intracellular signaling by introducing post-translational modifications to proteins and thereby altering their functions. The central nervous system (CNS) expresses different Nox isoforms during both development and adulthood. Here, we review the role of Nox-mediated ROS during CNS development. Specifically, we focus on how individual Nox isoforms contribute to signaling in neural stem cell maintenance and neuronal differentiation, as well as neurite outgrowth and guidance. We also discuss how ROS regulates the organization and dynamics of the actin cytoskeleton in the neuronal growth cone. Finally, we review recent evidence that Nox-derived ROS modulate axonal regeneration upon nervous system injury.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
16
|
Schiavone S, Tucci P, Trabace L, Morgese MG. Early Celastrol Administration Prevents Ketamine-Induced Psychotic-Like Behavioral Dysfunctions, Oxidative Stress and IL-10 Reduction in The Cerebellum of Adult Mice. Molecules 2019; 24:molecules24213993. [PMID: 31694174 PMCID: PMC6864687 DOI: 10.3390/molecules24213993] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
Administration of subanesthetic doses of ketamine during brain maturation represents a tool to mimic an early insult to the central nervous system (CNS). The cerebellum is a key player in psychosis pathogenesis, to which oxidative stress also contributes. Here, we investigated the impact of early celastrol administration on behavioral dysfunctions in adult mice that had received ketamine (30 mg/kg i.p.) at postnatal days (PNDs) 7, 9, and 11. Cerebellar levels of 8-hydroxydeoxyguanosine (8-OHdG), NADPH oxidase (NOX) 1 and NOX2, as well as of the calcium-binding protein parvalbumin (PV), were also assessed. Furthermore, celastrol effects on ketamine-induced alterations of proinflammatory (TNF-α, IL-6 and IL-1β) and anti-inflammatory (IL-10) cytokines in this brain region were evaluated. Early celastrol administration prevented ketamine-induced discrimination index decrease at adulthood. The same was found for locomotor activity elevations and increased close following and allogrooming, whereas no beneficial effects on sniffing impairment were detected. Ketamine increased 8-OHdG in the cerebellum of adult mice, which was also prevented by early celastrol injection. Cerebellar NOX1 levels were enhanced at adulthood following postnatal ketamine exposure. Celastrol per se induced NOX1 decrease in the cerebellum. This effect was more significant in animals that were early administered with ketamine. NOX2 levels did not change. Ketamine administration did not affect PV amount in the cerebellum. TNF-α levels were enhanced in ketamine-treated animals; however, this was not prevented by early celastrol administration. While no changes were observed for IL-6 and IL-1β levels, ketamine determined a reduction of cerebellar IL-10 expression, which was prevented by early celastrol treatment. Our results suggest that NOX inhibition during brain maturation prevents the development of psychotic-like behavioral dysfunctions, as well as the increased cerebellar oxidative stress and the reduction of IL-10 in the same brain region following ketamine exposure in postnatal life. This opens novel neuroprotective opportunities against early detrimental insults occurring during brain development.
Collapse
|
17
|
Mazzonetto PC, Ariza CB, Ocanha SG, de Souza TA, Ko GM, Menck CFM, Massironi SMG, Porcionatto MA. Mutation in NADPH oxidase 3 (NOX3) impairs SHH signaling and increases cerebellar neural stem/progenitor cell proliferation. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1502-1515. [PMID: 30853403 DOI: 10.1016/j.bbadis.2019.02.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 01/03/2023]
Abstract
Abnormalities in cerebellar structure and function may cause ataxia, a neurological dysfunction of motor coordination. In the course of the present study, we characterized a mutant mouse lineage with an ataxia-like phenotype. We localized the mutation on chromosome 17 and mapped it to position 1534 of the Nox3 gene, resulting in p.Asn64Tyr change. The primary defect observed in Nox3eqlb mice was increased proliferation of cerebellar granule cell precursors (GCPs). cDNA microarray comparing Nox3eqlb and BALB/c neonatal cerebellum revealed changes in the expression of genes involved in the control of cell proliferation. Nox3eqlb GCPs and NSC produce higher amounts of reactive oxygen species (ROS) and upregulate the expression of SHH target genes, such as Gli1-3 and Ccnd1 (CyclinD1). We hypothesize that this new mutation is responsible for an increase in proliferation via stimulation of the SHH pathway. We suggest this mutant mouse lineage as a new model to investigate the role of ROS in neuronal precursor cell proliferation.
Collapse
Affiliation(s)
- P C Mazzonetto
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - C B Ariza
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil; Department of General Pathology, Center of Biological Sciences, Universidade Estadual de Londrina (UEL), Brazil
| | - S G Ocanha
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - T A de Souza
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - G M Ko
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil
| | - C F M Menck
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - S M G Massironi
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo (USP), Brazil
| | - M A Porcionatto
- Department of Biochemistry, Laboratory of Neurobiology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Brazil.
| |
Collapse
|
18
|
Garza-Lombó C, Petrosyan P, Tapia-Rodríguez M, Valdovinos-Flores C, Gonsebatt ME. Systemic L-buthionine-S-R-sulfoximine administration modulates glutathione homeostasis via NGF/TrkA and mTOR signaling in the cerebellum. Neurochem Int 2018; 121:8-18. [PMID: 30300680 DOI: 10.1016/j.neuint.2018.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022]
Abstract
Glutathione (GSH) is an essential component of intracellular antioxidant systems that plays a primordial role in the protection of cells against oxidative stress, maintaining redox homeostasis and xenobiotic detoxification. GSH synthesis in the brain is limited by the availability of cysteine and glutamate. Cystine, the disulfide form of cysteine is transported into endothelial cells of the blood-brain barrier (BBB) and astrocytes via the system xc-, which is composed of xCT and the heavy chain of 4F2 cell surface antigen (4F2hc). Cystine is reduced inside the cells and the L-type amino acid transporter 1 (LAT1) transports cysteine from the endothelial cells into the brain, cysteine is transported into the neurons through the excitatory amino acid transporter 3 (EAAT3), also known as excitatory amino acid carrier 1 (EAAC1). The mechanistic/mammalian target of rapamycin (mTOR) and neurotrophins can activate signaling pathways that modulate amino acid transporters for GSH synthesis. The present study found that systemic L-buthionine-S-R-sulfoximine (BSO) administration selectively altered GSH homeostasis and EAAT3 levels in the mice cerebellum. Intraperitoneal treatment of mice with 6 mmol/kg of BSO depleted GSH and GSSG in the liver at 2 h of treatment. The cerebellum, but not other brain regions, exhibited a redox response. The mTOR and the neuronal growth factor (NGF)/tropomyosin receptor kinase A (TrkA) signaling pathways were activated and lead to an increase in the protein levels of the EAAT3 transporter, which was linked to an increase in the GSH/GSSG ratio and GSH concentration in the cerebellum at 0.5 and 2 h, respectively. Therefore, the cerebellum responds to peripheral GSH depletion via activation of the mTOR and NGF/TrkA pathways, which increase the transport of cysteine for GSH synthesis.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico.
| | - Pavel Petrosyan
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico.
| | - Miguel Tapia-Rodríguez
- Unidad de Microscopía, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico.
| | - Cesar Valdovinos-Flores
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico.
| | - María E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico.
| |
Collapse
|
19
|
Abstract
SIGNIFICANCE Numerous studies have demonstrated the actions of reactive oxygen species (ROS) as regulators of several physiological processes. In this study, we discuss how redox signaling mechanisms operate to control different processes such as neuronal differentiation, oligodendrocyte differentiation, dendritic growth, and axonal growth. Recent Advances: Redox homeostasis regulates the physiology of neural stem cells (NSCs). Notably, the neuronal differentiation process of NSCs is determined by a change toward oxidative metabolism, increased levels of mitochondrial ROS, increased activity of NADPH oxidase (NOX) enzymes, decreased levels of Nrf2, and differential regulation of different redoxins. Furthermore, during the neuronal maturation processes, NOX and MICAL produce ROS to regulate cytoskeletal dynamics, which control the dendritic and axonal growth, as well as the axonal guidance. CRITICAL ISSUES The redox homeostasis changes are, in part, attributed to cell metabolism and compartmentalized production of ROS, which is regulated, sensed, and transduced by different molecules such as thioredoxins, glutaredoxins, peroxiredoxins, and nucleoredoxin to control different signaling pathways in different subcellular regions. The study of how these elements cooperatively act is essential for the understanding of nervous system development, as well as the application of regenerative therapies that recapitulate these processes. FUTURE DIRECTIONS The information about these topics in the last two decades leads us to the conclusion that the role of ROS signaling in development of the nervous system is more important than it was previously believed and makes clear the importance of exploring in more detail the mechanisms of redox signaling. Antioxid. Redox Signal. 28, 1603-1625.
Collapse
Affiliation(s)
- Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular , Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
20
|
Rama Rao KV, Iring S, Younger D, Kuriakose M, Skotak M, Alay E, Gupta RK, Chandra N. A Single Primary Blast-Induced Traumatic Brain Injury in a Rodent Model Causes Cell-Type Dependent Increase in Nicotinamide Adenine Dinucleotide Phosphate Oxidase Isoforms in Vulnerable Brain Regions. J Neurotrauma 2018; 35:2077-2090. [PMID: 29648986 PMCID: PMC6098412 DOI: 10.1089/neu.2017.5358] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Blast-induced traumatic brain injury (bTBI) is a leading cause of morbidity in soldiers on the battlefield and in training sites with long-term neurological and psychological pathologies. Previous studies from our laboratory demonstrated activation of oxidative stress pathways after blast injury, but their distribution among different brain regions and their impact on the pathogenesis of bTBI have not been explored. The present study examined the protein expression of two isoforms: nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 and 2 (NOX1, NOX2), corresponding superoxide production, a downstream event of NOX activation, and the extent of lipid peroxidation adducts of 4-hydroxynonenal (4HNE) to a range of proteins. Brain injury was evaluated 4 h after the shock-wave exposure, and immunofluorescence signal quantification was performed in different brain regions. Expression of NOX isoforms displayed a differential increase in various brain regions: in hippocampus and thalamus, there was the highest increase of NOX1, whereas in the frontal cortex, there was the highest increase of NOX2 expression. Cell-specific analysis of changes in NOX expression with respect to corresponding controls revealed that blast resulted in a higher increase of NOX1 and NOX 2 levels in neurons compared with astrocytes and microglia. Blast exposure also resulted in increased superoxide levels in different brain regions, and such changes were reflected in 4HNE protein adduct formation. Collectively, this study demonstrates that primary blast TBI induces upregulation of NADPH oxidase isoforms in different regions of the brain parenchyma and that neurons appear to be at higher risk for oxidative damage compared with other neural cells.
Collapse
Affiliation(s)
- Kakulavarapu V Rama Rao
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Stephanie Iring
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Daniel Younger
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Matthew Kuriakose
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Maciej Skotak
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Eren Alay
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| | - Raj K Gupta
- 2 Department of Defense Blast Injury Research Program Coordinating Office, United States Army Medical Research and Materiel Command , Fort Detrick, Maryland
| | - Namas Chandra
- 1 Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology , Newark, New Jersey
| |
Collapse
|
21
|
nox2/cybb Deficiency Affects Zebrafish Retinotectal Connectivity. J Neurosci 2018; 38:5854-5871. [PMID: 29793976 DOI: 10.1523/jneurosci.1483-16.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 04/30/2018] [Accepted: 05/13/2018] [Indexed: 01/28/2023] Open
Abstract
NADPH oxidase (Nox)-derived reactive oxygen species (ROS) have been linked to neuronal polarity, axonal outgrowth, cerebellar development, regeneration of sensory axons, and neuroplasticity. However, the specific roles that individual Nox isoforms play during nervous system development in vivo remain unclear. To address this problem, we investigated the role of Nox activity in the development of retinotectal connections in zebrafish embryos. Zebrafish broadly express four nox genes (nox1, nox2/cybb, nox5, and duox) throughout the CNS during early development. Application of a pan-Nox inhibitor, celastrol, during the time of optic nerve (ON) outgrowth resulted in significant expansion of the ganglion cell layer (GCL), thinning of the ON, and a decrease in retinal axons reaching the optic tectum (OT). With the exception of GCL expansion, these effects were partially ameliorated by the addition of H2O2, a key ROS involved in Nox signaling. To address isoform-specific Nox functions, we used CRISPR/Cas9 to generate mutations in each zebrafish nox gene. We found that nox2/cybb chimeric mutants displayed ON thinning and decreased OT innervation. Furthermore, nox2/cybb homozygous mutants (nox2/cybb-/-) showed significant GCL expansion and mistargeted retinal axons in the OT. Neurite outgrowth from cultured zebrafish retinal ganglion cells was reduced by Nox inhibitors, suggesting a cell-autonomous role for Nox in these neurons. Collectively, our results show that Nox2/Cybb is important for retinotectal development in zebrafish.SIGNIFICANCE STATEMENT Most isoforms of NADPH oxidase (Nox) only produce reactive oxygen species (ROS) when activated by an upstream signal, making them ideal candidates for ROS signaling. Nox enzymes are present in neurons and their activity has been shown to be important for neuronal development and function largely by in vitro studies. However, whether Nox is involved in the development of axons and formation of neuronal connections in vivo has remained unclear. Using mutant zebrafish embryos, this study shows that a specific Nox isoform, Nox2/Cybb, is important for the establishment of axonal connections between retinal ganglion cells and the optic tectum.
Collapse
|
22
|
Metaplasticity in the Visual Cortex: Crosstalk Between Visual Experience and Reactive Oxygen Species. J Neurosci 2018; 38:5649-5665. [PMID: 29789380 DOI: 10.1523/jneurosci.2617-17.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 04/17/2018] [Accepted: 05/12/2018] [Indexed: 01/18/2023] Open
Abstract
Metaplasticity is the regulation of synaptic plasticity based on the history of previous synaptic activation. This concept was formulated after observing that synaptic changes in the visual cortex are not fixed, but dynamic and dependent on the history of visual information flux. In visual cortical neurons, sustained synaptic stimulation activate the enzymatic complex NOX2, resulting in the generation of reactive oxygen species (ROS). NOX2 is the main molecular structure responsible for translating neural activity into redox modulation of intracellular signaling pathways involved in plastic changes. Here, we studied the interaction between NOX2 and visual experience as metaplastic factors regulating synaptic plasticity at the supergranular layers of the mouse visual cortex. We found that genetic inhibition of NOX2 reverses the polarizing effects of dark rearing from LTP to LTD. In addition, we demonstrate that this process relies on changes in the NMDA receptor functioning. Altogether, this work indicates a role of ROS in the activity-dependent regulation of cortical synaptic plasticity.SIGNIFICANCE STATEMENT Synaptic plasticity in the visual cortex is modulated by the history of sensory experience and this modulation has been defined as metaplasticity. Dark rearing facilitates synaptic potentiation as a mechanism optimizing the range of synaptic modification. This process requires the production of reactive oxygen species mediated by the enzymatic complex NOX2. If the activity of NOX2 is inhibited, then visual deprivation results in synaptic depression. These findings increase our knowledge about metaplasticity and help in our understanding of how neural activity modulates cellular mechanisms of synaptic change.
Collapse
|
23
|
Nayernia Z, Colaianna M, Robledinos-Antón N, Gutzwiller E, Sloan-Béna F, Stathaki E, Hibaoui Y, Cuadrado A, Hescheler J, Stasia MJ, Saric T, Jaquet V, Krause KH. Decreased neural precursor cell pool in NADPH oxidase 2-deficiency: From mouse brain to neural differentiation of patient derived iPSC. Redox Biol 2017; 13:82-93. [PMID: 28575744 PMCID: PMC5454143 DOI: 10.1016/j.redox.2017.04.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 10/28/2022] Open
Abstract
There is emerging evidence for the involvement of reactive oxygen species (ROS) in the regulation of stem cells and cellular differentiation. Absence of the ROS-generating NADPH oxidase NOX2 in chronic granulomatous disease (CGD) patients, predominantly manifests as immune deficiency, but has also been associated with decreased cognition. Here, we investigate the role of NOX enzymes in neuronal homeostasis in adult mouse brain and in neural cells derived from human induced pluripotent stem cells (iPSC). High levels of NOX2 were found in mouse adult neurogenic regions. In NOX2-deficient mice, neurogenic regions showed diminished redox modifications, as well as decrease in neuroprecursor numbers and in expression of genes involved in neural differentiation including NES, BDNF and OTX2. iPSC from healthy subjects and patients with CGD were used to study the role of NOX2 in human in vitro neuronal development. Expression of NOX2 was low in undifferentiated iPSC, upregulated upon neural induction, and disappeared during neuronal differentiation. In human neurospheres, NOX2 protein and ROS generation were polarized within the inner cell layer of rosette structures. NOX2 deficiency in CGD-iPSCs resulted in an abnormal neural induction in vitro, as revealed by a reduced expression of neuroprogenitor markers (NES, BDNF, OTX2, NRSF/REST), and a decreased generation of mature neurons. Vector-mediated NOX2 expression in NOX2-deficient iPSCs rescued neurogenesis. Taken together, our study provides novel evidence for a regulatory role of NOX2 during early stages of neurogenesis in mouse and human.
Collapse
Affiliation(s)
- Zeynab Nayernia
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland
| | - Marilena Colaianna
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland
| | - Natalia Robledinos-Antón
- Instituto de Investigaciones Biomédicas "Alberto Sols", Faculty of Medicine, Autonomous University of Madrid (UAM), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eveline Gutzwiller
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland
| | - Frédérique Sloan-Béna
- Hôpitaux Universitaires de Genève HUG, Laboratoires de Cytogénétique Constitutionnelle, Service de Médecine Génétique, Geneva, Switzerland
| | - Elisavet Stathaki
- Hôpitaux Universitaires de Genève HUG, Laboratoires de Cytogénétique Constitutionnelle, Service de Médecine Génétique, Geneva, Switzerland
| | - Yousef Hibaoui
- Department of Genetic Medicine and Development, University of Geneva Medical School, 1 rue Michel Servet, 1211 Geneva, Switzerland
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas "Alberto Sols", Faculty of Medicine, Autonomous University of Madrid (UAM), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Marie-José Stasia
- Université Grenoble Alpes, Techniques de l'Ingénierie Médicale et de la Complexité- Grenoble, F38000 Grenoble, France
| | - Tomo Saric
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Vincent Jaquet
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva Medical School, 1-rue Michel Servet, 1211 Geneva, Switzerland.
| |
Collapse
|
24
|
Poulose SM, Rabin BM, Bielinski DF, Kelly ME, Miller MG, Thanthaeng N, Shukitt-Hale B. Neurochemical differences in learning and memory paradigms among rats supplemented with anthocyanin-rich blueberry diets and exposed to acute doses of 56Fe particles. LIFE SCIENCES IN SPACE RESEARCH 2017; 12:16-23. [PMID: 28212704 DOI: 10.1016/j.lssr.2016.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/04/2016] [Indexed: 06/06/2023]
Abstract
The protective effects of anthocyanin-rich blueberries (BB) on brain health are well documented and are particularly important under conditions of high oxidative stress, which can lead to "accelerated aging." One such scenario is exposure to space radiation, consisting of high-energy and -charge particles (HZE), which are known to cause cognitive dysfunction and deleterious neurochemical alterations. We recently tested the behavioral and neurochemical effects of acute exposure to HZE particles such as 56Fe, within 24-48h after exposure, and found that radiation primarily affects memory and not learning. Importantly, we observed that specific brain regions failed to upregulate antioxidant and anti-inflammatory mechanisms in response to this insult. To further examine these endogenous response mechanisms, we have supplemented young rats with diets rich in BB, which are known to contain high amounts of antioxidant-phytochemicals, prior to irradiation. Exposure to 56Fe caused significant neurochemical changes in hippocampus and frontal cortex, the two critical regions of the brain involved in cognitive function. BB supplementation significantly attenuated protein carbonylation, which was significantly increased by exposure to 56Fe in the hippocampus and frontal cortex. Moreover, BB supplementation significantly reduced radiation-induced elevations in NADPH-oxidoreductase-2 (NOX2) and cyclooxygenase-2 (COX-2), and upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) in the hippocampus and frontal cortex. Overall results indicate that 56Fe particles may induce their toxic effects on hippocampus and frontal cortex by reactive oxygen species (ROS) overload, which can cause alterations in the neuronal environment, eventually leading to hippocampal neuronal death and subsequent impairment of cognitive function. Blueberry supplementation provides an effective preventative measure to reduce the ROS load on the CNS in an event of acute HZE exposure.
Collapse
Affiliation(s)
- Shibu M Poulose
- USDA Human Nutrtion Research Center on Aging at Tufts University, 711 Washington Street, Boston MA 02111, USA
| | | | - Donna F Bielinski
- USDA Human Nutrtion Research Center on Aging at Tufts University, 711 Washington Street, Boston MA 02111, USA
| | - Megan E Kelly
- USDA Human Nutrtion Research Center on Aging at Tufts University, 711 Washington Street, Boston MA 02111, USA
| | - Marshall G Miller
- USDA Human Nutrtion Research Center on Aging at Tufts University, 711 Washington Street, Boston MA 02111, USA
| | - Nopporn Thanthaeng
- USDA Human Nutrtion Research Center on Aging at Tufts University, 711 Washington Street, Boston MA 02111, USA
| | - Barbara Shukitt-Hale
- USDA Human Nutrtion Research Center on Aging at Tufts University, 711 Washington Street, Boston MA 02111, USA.
| |
Collapse
|
25
|
Sun Z, Satomoto M, Adachi YU, Kinoshita H, Makita K. Inhibiting NADPH oxidase protects against long-term memory impairment induced by neonatal sevoflurane exposure in mice. Br J Anaesth 2016; 117:80-6. [PMID: 27147542 DOI: 10.1093/bja/aew064] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Neonatal exposure to anaesthetics such as sevoflurane has been reported to result in behavioural deficits in rodents. However, while oxidative injury is thought to play an underlying pathological role, the mechanisms of neurotoxicity remain unclear. In the present study, we investigated whether the NADPH oxidase inhibitor apocynin protects against long-term memory impairment produced by neonatal sevoflurane exposure in mice. METHODS Postnatal day six mice were divided into four groups; (1) non-anaesthesia, (2) intraperitoneal apocynin (50 mg kg(-1)) treatment, (3) 3% sevoflurane exposure for 6 h, and (4) apocynin treatment combined with sevoflurane exposure. Superoxide concentrations and NADPH oxidase expression in the brain were determined using dihydroethidium fluorescence and immunoblotting, respectively. Cleaved caspase-3 immunoblotting was used for the detection of apoptosis, and cytochrome c immunoblotting was performed to evaluate mitochondrial function. Long-term cognitive impairment was evaluated using the fear conditioning test in adulthood. RESULTS Sevoflurane exposure increased concentrations of superoxide (109%) and the NADPH oxidase subunit p22phox (39%) in the brain, and apocynin abolished these increases. Neonatal sevoflurane exposure caused learning deficits in adulthood. Apocynin also maintained long-term memory function in mice given neonatal sevoflurane exposure, and it reduced apoptosis and decreased cytochrome c concentrations in the brains of these mice. CONCLUSIONS Apocynin reduces neuronal apoptosis and protects against long-term memory impairment in mice, neonatally exposed to sevoflurane by reducing superoxide concentrations. These findings suggest that NADPH oxidase inhibitors may protect against cognitive dysfunction resulting from neonatal anaesthesia.
Collapse
Affiliation(s)
- Z Sun
- Department of Anaesthesiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - M Satomoto
- Department of Anaesthesiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Y U Adachi
- Department of Anaesthesiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - H Kinoshita
- Department of Anaesthesiology, Aichi Medical University School of Medicine, Aichi, Japan
| | - K Makita
- Department of Anaesthesiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
26
|
Amano I, Takatsuru Y, Toya S, Haijima A, Iwasaki T, Grasberger H, Refetoff S, Koibuchi N. Aberrant Cerebellar Development in Mice Lacking Dual Oxidase Maturation Factors. Thyroid 2016; 26:741-52. [PMID: 26914863 PMCID: PMC4860669 DOI: 10.1089/thy.2015.0034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Thyroid hormone (TH) plays a key role in the developing brain, including the cerebellum. TH deficiency induces organizational changes of the cerebellum, causing cerebellar ataxia. However, the mechanisms causing these abnormalities are poorly understood. Various animal models have been used to study the mechanism. Lacking dual oxidase (DUOX) and its maturation factor (DUOXA) are major inducers of congenital hypothyroidism. Thus, this study examined the organizational changes of the cerebellum using knockout mice of the Duoxa gene (Duoxa-/-). METHODS The morphological, behavioral, and electrophysiological changes were analyzed in wild type (Wt) and Duoxa-deficient (Duoxa-/-) mice from postnatal day (P) 10 to P30. To detect the changes in the expression levels of presynaptic proteins, Western blot analysis was performed. RESULTS The proliferation and migration of granule cells was delayed after P15 in Duoxa-/- mice. However, these changes disappeared by P25. Although the cerebellar structure of Duoxa-/- mice was not significantly different from that of Wt mice at P25, motor coordination was impaired. It was also found that the amplitude of paired-pulse facilitation at parallel fiber-Purkinje cell synapses decreased in Duoxa-/- mice, particularly at P15. There were no differences between expression levels of presynaptic proteins regulating neurotransmitter release at P25. CONCLUSIONS These results indicate that the anatomical catch-up growth of the cerebellum did not normalize its function because of the disturbance of neuronal circuits by the combined effect of hypothyroidism and functional disruption of the DUOX/DUOXA complex.
Collapse
Affiliation(s)
- Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yusuke Takatsuru
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Syutaro Toya
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Asahi Haijima
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Toshiharu Iwasaki
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Helmut Grasberger
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
- Department of Genetics, The University of Chicago, Chicago, Illinois
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
27
|
Satomoto M, Makita K. Anesthesia-induced neurotoxicity in an animal model of the developing brain: mechanism and therapies. Neural Regen Res 2016; 11:1407-1408. [PMID: 27857736 PMCID: PMC5090835 DOI: 10.4103/1673-5374.191207] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Maiko Satomoto
- Department of Anesthesiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koshi Makita
- Department of Anesthesiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
28
|
Weaver CJ, Leung YF, Suter DM. Expression dynamics of NADPH oxidases during early zebrafish development. J Comp Neurol 2015; 524:2130-41. [PMID: 26662995 DOI: 10.1002/cne.23938] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/03/2015] [Accepted: 11/24/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Cory J. Weaver
- Department of Biological Sciences; Purdue University; West Lafayette Indiana 47907
| | - Yuk Fai Leung
- Department of Biological Sciences; Purdue University; West Lafayette Indiana 47907
| | - Daniel M. Suter
- Department of Biological Sciences; Purdue University; West Lafayette Indiana 47907
- Bindley Bioscience Center; Purdue University; West Lafayette Indiana 47907
| |
Collapse
|
29
|
Peroxynitrite is Involved in the Apoptotic Death of Cultured Cerebellar Granule Neurons Induced by Staurosporine, but not by Potassium Deprivation. Neurochem Res 2015; 41:316-27. [PMID: 26700430 DOI: 10.1007/s11064-015-1805-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 01/13/2023]
Abstract
Nitric oxide (NO) regulates numerous physiological process and is the main source of reactive nitrogen species (RNS). NO promotes cell survival, but it also induces apoptotic death having been involved in the pathogenesis of several neurodegenerative diseases. NO and superoxide anion react to form peroxynitrite, which accounts for most of the deleterious effects of NO. The mechanisms by which these molecules regulate the apoptotic process are not well understood. In this study, we evaluated the role of NO and peroxynitrite in the apoptotic death of cultured cerebellar granule neurons (CGN), which are known to experience apoptosis by staurosporine (St) or potassium deprivation (K5). We found that CGN treated with the peroxynitrite catalyst, FeTTPs were completely rescued from St-induced death, but not from K5-induced death. On the other hand, the inhibition of the inducible nitric oxide synthase partially protected cell viability in CGN treated with K5, but not with St, while the inhibitor L-NAME further reduced the cell viability in St, but it did not affect K5. Finally, an inhibitor of the soluble guanylate cyclase (sGC) diminished the cell viability in K5, but not in St. Altogether, these results shows that NO promotes cell survival in K5 through sGC-cGMP and promotes cell death by other mechanisms, while in St NO promotes cell survival independently of cGMP and peroxynitrite results critical for St-induced death. Our results suggest that RNS are differentially handled by CGN during cell death depending on the death-inducing conditions.
Collapse
|
30
|
Reactive Oxygen Species in Planarian Regeneration: An Upstream Necessity for Correct Patterning and Brain Formation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:392476. [PMID: 26180588 PMCID: PMC4477255 DOI: 10.1155/2015/392476] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 04/30/2015] [Accepted: 05/04/2015] [Indexed: 12/12/2022]
Abstract
Recent research highlighted the impact of ROS as upstream regulators of tissue regeneration. We investigated their role and targeted processes during the regeneration of different body structures using the planarian Schmidtea mediterranea, an organism capable of regenerating its entire body, including its brain. The amputation of head and tail compartments induces a ROS burst at the wound site independently of the orientation. Inhibition of ROS production by diphenyleneiodonium (DPI) or apocynin (APO) causes regeneration defaults at both the anterior and posterior wound sites, resulting in reduced regeneration sites (blastemas) and improper tissue homeostasis. ROS signaling is necessary for early differentiation and inhibition of the ROS burst results in defects on the regeneration of the nervous system and on the patterning process. Stem cell proliferation was not affected, as indicated by histone H3-P immunostaining, fluorescence-activated cell sorting (FACS), in situ hybridization of smedwi-1, and transcript levels of proliferation-related genes. We showed for the first time that ROS modulate both anterior and posterior regeneration in a context where regeneration is not limited to certain body structures. Our results indicate that ROS are key players in neuroregeneration through interference with the differentiation and patterning processes.
Collapse
|
31
|
Olguín-Albuerne M, Morán J. ROS produced by NOX2 control in vitro development of cerebellar granule neurons development. ASN Neuro 2015; 7:7/2/1759091415578712. [PMID: 25873309 PMCID: PMC4720178 DOI: 10.1177/1759091415578712] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Reactive oxygen species (ROS) act as signaling molecules that regulate nervous system physiology. ROS have been related to neural differentiation, neuritogenesis, and programmed cell death. Nevertheless, little is known about the mechanisms involved in the regulation of ROS during neuronal development. In this study, we evaluated the mechanisms by which ROS are regulated during neuronal development and the implications of these molecules in this process. Primary cultures of cerebellar granule neurons (CGN) were used to address these issues. Our results show that during the first 3 days of CGN development in vitro (days in vitro; DIV), the levels of ROS increased, reaching a peak at 2 and 3 DIV under depolarizing (25 mM KCl) and nondepolarizing (5 mM KCl) conditions. Subsequently, under depolarizing conditions, the ROS levels markedly decreased, but in nondepolarizing conditions, the ROS levels increased gradually. This correlated with the extent of CGN maturation. Also, antioxidants and NADPH-oxidases (NOX) inhibitors reduced the expression of Tau and MAP2. On the other hand, the levels of glutathione markedly increased at 1 DIV. We inferred that the ROS increase at this time is critical for cell survival because glutathione depletion leads to axonal degeneration and CGN death only at 2 DIV. During the first 3 DIV, NOX2 was upregulated and expressed in filopodia and growth cones, which correlated with the hydrogen peroxide (H2O2) distribution in the cell. Finally, NOX2 KO CGN showed shorter neurites than wild-type CGN. Taken together, these results suggest that the regulation of ROS is critical during the early stages of CGN development.
Collapse
Affiliation(s)
- Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| |
Collapse
|
32
|
Poulose SM, Bielinski DF, Carrihill-Knoll KL, Rabin BM, Shukitt-Hale B. Protective effects of blueberry- and strawberry diets on neuronal stress following exposure to 56Fe particles. Brain Res 2014; 1593:9-18. [DOI: 10.1016/j.brainres.2014.10.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
|
33
|
Abstract
Reactive oxygen species (ROS) are signaling factors involved in many intracellular transduction pathways. In the nervous system, ROS are thought to modulate various mechanisms of synaptic plasticity. One important source of ROS production in the brain is the NADPH oxidase complex. Stimulation of NMDA receptors activates NADPH oxidase, which provides selective oxidative responses accompanying the induction of synaptic changes. The activity of NADPH oxidase is known to be crucial for the induction of LTP in the hippocampus. However, the involvement of this complex in cortical synaptic plasticity is still unclear. Here we provide evidence that genetic ablation of NOX2 (the prototypical member of NADPH oxidase family of proteins) suppresses LTP and LTD in the primary visual cortex of the mouse. We also found that the involvement of NOX2 on LTP is partially age-dependent, as the activity of this complex is not critical for mechanisms of synaptic potentiation occurring in immature animals. Furthermore, we show that inhibition of NOX2 reduces the NMDA receptor function, suggesting a possible mechanism that could be the basis of the effects on synaptic plasticity.
Collapse
|
34
|
Abstract
SIGNIFICANCE There is increasing evidence that the generation of reactive oxygen species (ROS) in the central nervous system (CNS) involves the NOX family of nicotinamide adenine dinucleotide phosphate oxidases. Controlled ROS generation appears necessary for optimal functioning of the CNS through fine-tuning of redox-sensitive signaling pathways, while overshooting ROS generation will lead to oxidative stress and CNS disease. RECENT ADVANCES NOX enzymes are not only restricted to microglia (i.e. brain phagocytes) but also expressed in neurons, astrocytes, and the neurovascular system. NOX enzymes are involved in CNS development, neural stem cell biology, and the function of mature neurons. While NOX2 appears to be a major source of pathological oxidative stress in the CNS, other NOX isoforms might also be of importance, for example, NOX4 in stroke. Globally speaking, there is now convincing evidence for a role of NOX enzymes in various neurodegenerative diseases, cerebrovascular diseases, and psychosis-related disorders. CRITICAL ISSUES The relative importance of specific ROS sources (e.g., NOX enzymes vs. mitochondria; NOX2 vs. NOX4) in different pathological processes needs further investigation. The absence of specific inhibitors limits the possibility to investigate specific therapeutic strategies. The uncritical use of non-specific inhibitors (e.g., apocynin, diphenylene iodonium) and poorly validated antibodies may lead to misleading conclusions. FUTURE DIRECTIONS Physiological and pathophysiological studies with cell-type-specific knock-out mice will be necessary to delineate the precise functions of NOX enzymes and their implications in pathomechanisms. The development of CNS-permeant, specific NOX inhibitors will be necessary to advance toward therapeutic applications.
Collapse
Affiliation(s)
- Zeynab Nayernia
- 1 Department of Pathology and Immunology, Geneva Medical Faculty, Geneva University Hospitals, Centre Médical Universitaire , Geneva, Switzerland
| | | | | |
Collapse
|
35
|
Salucci S, Ambrogini P, Lattanzi D, Betti M, Gobbi P, Galati C, Galli F, Cuppini R, Minelli A. Maternal dietary loads of alpha-tocopherol increase synapse density and glial synaptic coverage in the hippocampus of adult offspring. Eur J Histochem 2014; 58:2355. [PMID: 24998923 PMCID: PMC4083323 DOI: 10.4081/ejh.2014.2355] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/11/2014] [Accepted: 03/12/2014] [Indexed: 01/12/2023] Open
Abstract
An increased intake of the antioxidant α-Tocopherol (vitamin E) is recommended in complicated pregnancies, to prevent free radical damage to mother and fetus. However, the anti-PKC and antimitotic activity of α-Tocopherol raises concerns about its potential effects on brain development. Recently, we found that maternal dietary loads of α-Tocopherol through pregnancy and lactation cause developmental deficit in hippocampal synaptic plasticity in rat offspring. The defect persisted into adulthood, with behavioral alterations in hippocampus-dependent learning. Here, using the same rat model of maternal supplementation, ultrastructural morphometric studies were carried out to provide mechanistic interpretation to such a functional impairment in adult offspring by the occurrence of long-term changes in density and morphological features of hippocampal synapses. Higher density of axo-spinous synapses was found in CA1 stratum radiatum of α-Tocopherol-exposed rats compared to controls, pointing to a reduced synapse pruning. No morphometric changes were found in synaptic ultrastructural features, i.e., perimeter of axon terminals, length of synaptic specializations, extension of bouton-spine contact. Glia-synapse anatomical relationship was also affected. Heavier astrocytic coverage of synapses was observed in Tocopherol-treated offspring, notably surrounding axon terminals; moreover, the percentage of synapses contacted by astrocytic endfeet at bouton-spine interface (tripartite synapses) was increased. These findings indicate that gestational and neonatal exposure to supranutritional tocopherol intake can result in anatomical changes of offspring hippocampus that last through adulthood. These include a surplus of axo-spinous synapses and an aberrant glia-synapse relationship, which may represent the morphological signature of previously described alterations in synaptic plasticity and hippocampus-dependent learning.
Collapse
|
36
|
Zucker SN, Fink EE, Bagati A, Mannava S, Bianchi-Smiraglia A, Bogner PN, Wawrzyniak JA, Foley C, Leonova KI, Grimm MJ, Moparthy K, Ionov Y, Wang J, Liu S, Sexton S, Kandel ES, Bakin AV, Zhang Y, Kaminski N, Segal BH, Nikiforov MA. Nrf2 amplifies oxidative stress via induction of Klf9. Mol Cell 2014; 53:916-928. [PMID: 24613345 DOI: 10.1016/j.molcel.2014.01.033] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 12/19/2013] [Accepted: 01/28/2014] [Indexed: 12/25/2022]
Abstract
Reactive oxygen species (ROS) activate NF-E2-related transcription factor 2 (Nrf2), a key transcriptional regulator driving antioxidant gene expression and protection from oxidant injury. Here, we report that in response to elevation of intracellular ROS above a critical threshold, Nrf2 stimulates expression of transcription Kruppel-like factor 9 (Klf9), resulting in further Klf9-dependent increases in ROS and subsequent cell death. We demonstrated that Klf9 independently causes increased ROS levels in various types of cultured cells and in mouse tissues and is required for pathogenesis of bleomycin-induced pulmonary fibrosis in mice. Mechanistically, Klf9 binds to the promoters and alters the expression of several genes involved in the metabolism of ROS, including suppression of thioredoxin reductase 2, an enzyme participating in ROS clearance. Our data reveal an Nrf2-dependent feedforward regulation of ROS and identify Klf9 as a ubiquitous regulator of oxidative stress and lung injury.
Collapse
Affiliation(s)
- Shoshanna N Zucker
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Emily E Fink
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Archis Bagati
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Sudha Mannava
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Paul N Bogner
- Department of Pathology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Joseph A Wawrzyniak
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Colleen Foley
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Katerina I Leonova
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Melissa J Grimm
- Department of Medicine Immunology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Kalyana Moparthy
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Yurij Ionov
- Department of Cancer Genetics, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Sandra Sexton
- Department of Laboratory Animal Resources, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Eugene S Kandel
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Andrei V Bakin
- Department of Cancer Genetics, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Yuesheng Zhang
- Department of Cancer Prevention and Control, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Naftali Kaminski
- Roswell Park Cancer Institute, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Brahm H Segal
- Department of Medicine Immunology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| | - Mikhail A Nikiforov
- Department of Cell Stress Biology, Buffalo, New York, 14263, USA, Department of Internal Medicine, Yale University, New Haven, Connecticut 06520 USA
| |
Collapse
|
37
|
Martín MV, Distefano AM, Zabaleta EJ, Pagnussat GC. New insights into the functional roles of reactive oxygen species during embryo sac development and fertilization in Arabidopsis thaliana. PLANT SIGNALING & BEHAVIOR 2013; 8:doi: 10.4161/psb.25714. [PMID: 23887494 PMCID: PMC4091057 DOI: 10.4161/psb.25714] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Previously considered as toxic by-products of aerobic metabolism, reactive oxygen species (ROS) are emerging as essential signaling molecules in eukaryotes. Recent evidence showed that maintenance of ROS homeostasis during female gametophyte development is crucial for embryo sac patterning and fertilization. Although ROS are exclusively detected in the central cell of mature embryo sacs, the study of mutants deficient in ROS homeostasis suggests that controlled oxidative bursts might take place earlier during gametophyte development. Also, a ROS burst that depends on pollination takes place inside the embryo sac. This oxidative response might be required for pollen tube growth arrest and for sperm cell release. In this mini-review, we will focus on new insights into the role of ROS during female gametophyte development and fertilization. Special focus will be made on the mitochondrial Mn-Superoxide dismutase (MSD1), which has been recently reported to be essential for maintaining ROS homeostasis during embryo sac formation.
Collapse
|