1
|
Ergul Erkec O, Acikgoz E, Huyut Z, Akyol ME, Ozyurt EO, Keskin S. Ghrelin ameliorates neuronal damage, oxidative stress, inflammatory parameters, and GFAP expression in traumatic brain injury. Brain Inj 2024; 38:514-523. [PMID: 38433464 DOI: 10.1080/02699052.2024.2324012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE This study investigated the effects of ghrelin on oxidative stress, working memory, inflammatory parameters, and neuron degeneration. METHODS TBI was produced with the weight-drop technique. Rats in the G+TBI and TBI+G groups received ghrelin for 7 or 2 days, respectively. The control group received saline. On the 8th day of the study, the brain and blood tissue were taken under anesthesia. RESULTS A significant increase in brain GSH-PX, MDA, IL-1β, TGF-β1, and IL-8 levels and a significant decrease in CAT levels were found in the TBI group compared to the control. Serum MDA, GSH, IL-1β, and IL-8 levels were increased with TBI. Ghrelin treatment after TBI significantly increased the serum GSH, CAT, GSH-PX, and brain GSH and CAT levels, while it significantly decreased the serum MDA, IL-1β, and brain MDA, TGF-β1, and IL-8 levels. Histological evaluations revealed that ghrelin treatment led to a reduction in inflammation, while also significantly ameliorating TBI-induced neuron damage and vascular injuries. Immunohistochemistry staining showed that GFAP staining intensity was significantly increased in the cortex and hippocampus in TBI, and GFAP immunoreactivity was decreased with ghrelin treatment. CONCLUSION The results from this study suggested that ghrelin may have curative effects on TBI.
Collapse
Affiliation(s)
- Ozlem Ergul Erkec
- Department of Physiology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Mehmet Edip Akyol
- Department of Neurosurgery, Faculty of Mecine, Van Yuzuncu Yil University, Van, Turkey
| | | | - Sıddık Keskin
- Department of Biostatistics, Van Yuzuncu Yıl University, Van, Turkey
| |
Collapse
|
2
|
Ohashi SN, DeLong JH, Kozberg MG, Mazur-Hart DJ, van Veluw SJ, Alkayed NJ, Sansing LH. Role of Inflammatory Processes in Hemorrhagic Stroke. Stroke 2023; 54:605-619. [PMID: 36601948 DOI: 10.1161/strokeaha.122.037155] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Hemorrhagic stroke is the deadliest form of stroke and includes the subtypes of intracerebral hemorrhage and subarachnoid hemorrhage. A common cause of hemorrhagic stroke in older individuals is cerebral amyloid angiopathy. Intracerebral hemorrhage and subarachnoid hemorrhage both lead to the rapid collection of blood in the central nervous system and generate inflammatory immune responses that involve both brain resident and infiltrating immune cells. These responses are complex and can contribute to both tissue recovery and tissue injury. Despite the interconnectedness of these major subtypes of hemorrhagic stroke, few reviews have discussed them collectively. The present review provides an update on inflammatory processes that occur in response to intracerebral hemorrhage and subarachnoid hemorrhage, and the role of inflammation in the pathophysiology of cerebral amyloid angiopathy-related hemorrhage. The goal is to highlight inflammatory processes that underlie disease pathology and recovery. We aim to discuss recent advances in our understanding of these conditions and identify gaps in knowledge with the potential to develop effective therapeutic strategies.
Collapse
Affiliation(s)
- Sarah N Ohashi
- Department of Neurology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
- Department of Immunobiology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
| | - Jonathan H DeLong
- Department of Neurology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
- Department of Immunobiology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
| | - Mariel G Kozberg
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital/ Harvard Medical School, Boston (M.G.K., S.J.v.V.)
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown (M.G.K., S.J.v.V.)
| | - David J Mazur-Hart
- Department of Neurological Surgery (D.J.M.-H.), Oregon Health and Science University (OHSU), Portland
| | - Susanne J van Veluw
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital/ Harvard Medical School, Boston (M.G.K., S.J.v.V.)
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown (M.G.K., S.J.v.V.)
| | - Nabil J Alkayed
- Department of Anesthesiology & Perioperative Medicine and Knight Cardiovascular Institute (N.J.A.), Oregon Health and Science University (OHSU), Portland
| | - Lauren H Sansing
- Department of Neurology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
- Department of Immunobiology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
| |
Collapse
|
3
|
Song D, Yeh CT, Wang J, Guo F. Perspectives on the mechanism of pyroptosis after intracerebral hemorrhage. Front Immunol 2022; 13:989503. [PMID: 36131917 PMCID: PMC9484305 DOI: 10.3389/fimmu.2022.989503] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/17/2022] [Indexed: 12/18/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a highly harmful neurological disorder with high rates of mortality, disability, and recurrence. However, effective therapies are not currently available. Secondary immune injury and cell death are the leading causes of brain injury and a poor prognosis. Pyroptosis is a recently discovered form of programmed cell death that differs from apoptosis and necrosis and is mediated by gasdermin proteins. Pyroptosis is caused by multiple pathways that eventually form pores in the cell membrane, facilitating the release of inflammatory substances and causing the cell to rupture and die. Pyroptosis occurs in neurons, glial cells, and endothelial cells after ICH. Furthermore, pyroptosis causes cell death and releases inflammatory factors such as interleukin (IL)-1β and IL-18, leading to a secondary immune-inflammatory response and further brain damage. The NOD-like receptor protein 3 (NLRP3)/caspase-1/gasdermin D (GSDMD) pathway plays the most critical role in pyroptosis after ICH. Pyroptosis can be inhibited by directly targeting NLRP3 or its upstream molecules, or directly interfering with caspase-1 expression and GSDMD formation, thus significantly improving the prognosis of ICH. The present review discusses key pathological pathways and regulatory mechanisms of pyroptosis after ICH and suggests possible intervention strategies to mitigate pyroptosis and brain dysfunction after ICH.
Collapse
Affiliation(s)
- Dengpan Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Fuyou Guo, ; Jian Wang, ; Chi-Tai Yeh,
| |
Collapse
|
4
|
Witsch J, Spalart V, Martinod K, Schneider H, Oertel J, Geisel J, Hendrix P, Hemmer S. Neutrophil Extracellular Traps and Delayed Cerebral Ischemia in Aneurysmal Subarachnoid Hemorrhage. Crit Care Explor 2022; 4:e0692. [PMID: 35620772 PMCID: PMC9116951 DOI: 10.1097/cce.0000000000000692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
IMPORTANCE Myeloperoxidase (MPO)-DNA complexes, biomarkers of neutrophil extracellular traps (NETs), have been associated with arterial and venous thrombosis. Their role in aneurysmal subarachnoid hemorrhage (aSAH) is unknown. OBJECTIVES To assess whether serum MPO-DNA complexes are present in patients with aSAH and whether they are associated with delayed cerebral ischemia (DCI). DESIGN SETTING AND PARTICIPANTS Post-hoc analysis of a prospective, observational single-center study, with de novo serum biomarker measurements in consecutive patients with aSAH between July 2018 and September 2020, admitted to a tertiary care neuroscience ICU. MAIN OUTCOMES AND MEASURES We analyzed serum obtained at admission and hospital day 4 for concentrations of MPO-DNA complexes. The primary outcome was DCI, defined as new infarction on brain CT. The secondary outcome was clinical vasospasm, a composite of clinical and transcranial Doppler parameters. We used Wilcoxon signed-rank-test to assess for differences between paired measures. RESULTS Among 100 patients with spontaneous subarachnoid hemorrhage, mean age 59 years (sd ± 13 yr), 55% women, 78 had confirmed aSAH. Among these, 29 (37%) developed DCI. MPO-DNA complexes were detected in all samples. The median MPO-DNA level was 33 ng/mL (interquartile range [IQR], 18-43 ng/mL) at admission, and 22 ng/mL (IQR, 11-31 ng/mL) on day 4 (unpaired test; p = 0.015). We found a significant reduction in MPO-DNA levels from admission to day 4 in patients with DCI (paired test; p = 0.036) but not in those without DCI (p = 0.17). There was a similar reduction in MPO-DNA levels between admission and day 4 in patients with (p = 0.006) but not in those without clinical vasospasm (p = 0.47). CONCLUSIONS AND RELEVANCE This is the first study to detect the NET biomarkers MPO-DNA complexes in peripheral serum of patients with aSAH and to associate them with DCI. A pronounced reduction in MPO-DNA levels might serve as an early marker of DCI. This diagnostic potential of MPO-DNA complexes and their role as potential therapeutic targets in aSAH should be explored further.
Collapse
Affiliation(s)
- Jens Witsch
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Valérie Spalart
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Hauke Schneider
- Department of Neurology, University Hospital Augsburg, Augsburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Jürgen Geisel
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Medical Center, Homburg/Saar, Germany
| | - Philipp Hendrix
- Department of Neurosurgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Sina Hemmer
- Department of Neurosurgery, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
5
|
Tang J, Hu L, Long F, Zhang J, Tang J. Action mechanism of early cerebral injuries after spontaneous subarachnoid hemorrhage by silence Ghrelin and angiogenic factor with G-patch and FHA domain 1. Bioengineered 2022; 13:7340-7350. [PMID: 35259055 PMCID: PMC8974202 DOI: 10.1080/21655979.2022.2037373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The objective of the research was to investigate action mechanism of oxidative stress and cerebral injuries after subarachnoid hemorrhage (SAH) by Ghrelin and angiogenic factor G-patch and FHA domain 1 (Aggf1) and offer new research ideas to SAH clinical treatment and SAH-induced early cerebral injuries. SAH rat models were prepared by prechiasmatic anterior cistern injection. Specific Ghrelin and Aggf1 small interfering ribonucleic acid (siRNA) were designed and injected into silence Ghrelin or Aggf1 in rat left lateral ventricles. Rats were divided randomly into sham-operated (sham), SAH model, negative control siRNA, Ghrelin silence (Ghrelin(-/-)), and Aggf1 silence groups. Changes of rat neurological impairment, encephaledema, cerebral tissue phosphorylated protein kinase (p-Akt), and content changes of caspase-3 protein and oxidative stress indexes were observed, including glutathione (GSH) and oxidized glutathione (GSSG). Results showed scores of neurological impairment and water content in SAH model group were reduced compared with sham group, while p-Akt protein and GSH contents were enhanced. However, caspase-3 protein and GSSG contents were declined, showing statistically meaningful difference (P < 0.05). Compared with SAH model group, scores of neurological impairment, cerebral tissue water content, and caspase-3 protein and GSSG contents in silence Ghrelin and Aggf1 groups were increased, while p-Akt protein and GSH contents were decreased, demonstrating statistically meaningful difference (P < 0.05). To conclude, silence Ghrelin and Aggf1 aggravated early cerebral injuries after SAH, revealing that Ghrelin and Aggf1 could protect brains to some degree.
Collapse
Affiliation(s)
- Jianxun Tang
- Department of Cerebrovascular Disease, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ligang Hu
- Department of Cerebrovascular Disease, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Feng Long
- Department of Cerebrovascular Disease, General Medical College of Guilin Medical College, Guilin, China
| | - Jie Zhang
- Department of Cerebrovascular Disease, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jingfeng Tang
- Department of Cerebrovascular Disease, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
6
|
Tamer SA, Koyuncuoğlu T, Karagöz A, Akakın D, Yüksel M, Yeğen BÇ. Nesfatin-1 ameliorates oxidative brain damage and memory impairment in rats induced with a single acute epileptic seizure. Life Sci 2022; 294:120376. [DOI: 10.1016/j.lfs.2022.120376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 12/20/2022]
|
7
|
A 10-day mild treadmill exercise performed before an epileptic seizure alleviates oxidative injury in the skeletal muscle and brain tissues of the rats. MARMARA MEDICAL JOURNAL 2022. [DOI: 10.5472/marumj.1056192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
8
|
Ferrer B, Suresh H, Tinkov AA, Santamaria A, Rocha JB, Skalny AV, Bowman AB, Aschner M. Ghrelin attenuates methylmercury-induced oxidative stress in neuronal cells. Mol Neurobiol 2022; 59:2098-2115. [PMID: 35040042 DOI: 10.1007/s12035-022-02726-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/30/2021] [Indexed: 11/28/2022]
Abstract
Methylmercury (MeHg) is a global pollutant, which can cause damage to the central nervous system at both high-acute and chronic-low exposures, especially in vulnerable populations, such as children and pregnant women. Nowadays, acute-high poisoning is rare. However, chronic exposure to low MeHg concentrations via fish consumption remains a health concern. Current therapeutic strategies for MeHg poisoning are based on the use of chelators. However, these therapies have limited efficacy. Ghrelin is a gut hormone with an important role in regulating physiologic processes. It has been reported that ghrelin plays a protective role against the toxicity of several xenobiotics. Here, we explored the role of ghrelin as a putative protector against MeHg-induced oxidative stress. Our data show that ghrelin was able to ameliorate MeHg-induced reactive oxygen species (ROS) production in primary neuronal hypothalamic and hippocampal cultures. An analogous effect was observed in mouse hypothalamic neuronal GT 1-7 cells. Using this model, our novel findings show that antioxidant protection of ghrelin against MeHg is mediated by glutathione upregulation and induction of the NRF2/NQO1 pathway.
Collapse
Affiliation(s)
- Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| | - Harshini Suresh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University, Moscow, Russia.,Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, Russia
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular Y Nanotecnología, Instituto Nacional de Neurología Y Neurocirugía, 14269, Mexico City, Mexico
| | - João Batista Rocha
- Departamento de Bioquímica E Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Anatoly V Skalny
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", IM Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia.,Department of Bioelementology, KG Razumovsky Moscow State University of Technologies and Management, Moscow, Russia
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA. .,Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
9
|
Badi R. Acylated Ghrelin Attenuates l-Thyroxin-induced Cardiac Damage in Rats by Antioxidant and Anti-inflammatory Effects and Downregulating Components of the Cardiac Renin-angiotensin System. J Cardiovasc Pharmacol 2021; 78:422-436. [PMID: 34132689 DOI: 10.1097/fjc.0000000000001084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/26/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT This study investigated the protective effect of acylated ghrelin (AG) against l-thyroxin (l-Thy)-induced cardiac damage in rats and examined possible mechanisms. Male rats were divided into five intervention groups of 12 rats/group: control, control + AG, l-Thy, l-Thy + AG, and l-Thy + AG + [D-Lys3]-GHRP-6 (AG antagonist). l-Thy significantly reduced the levels of AG and des-acyl ghrelin and the AG to des-acyl ghrelin ratio. Administration of AG to l-Thy-treated rats reduced cardiac weights and levels of reactive oxygen species and preserved the function and structure of the left ventricle. In addition, AG also reduced the protein levels of cleaved caspase-3 and cytochrome c and prevented mitochondrial permeability transition pore opening. In the left ventricle of both control + AG-treated and l-Thy + AG-treated rats, AG significantly increased left ventricular levels of manganese superoxide dismutase (SOD2), total glutathione (GSH), and Bcl2. It also reduced the levels of malondialdehyde, tumor necrosis factor-α (TNF-α), interleukin-6, and Bax and the nuclear activity of nuclear factor-kappa B. Concomitantly, in both treated groups, AG reduced the mRNA and protein levels of NADPH oxidase 1, angiotensin (Ang) II type 1 receptor, and Ang-converting enzyme 2. All the beneficial effects of AG in l-Thy-treated rats were prevented by the coadministration of [D-Lys3]-GHRP-6, a selective growth hormone secretagogue receptor subtype 1a antagonist. In conclusion, AG protects against hyperthyroidism-induced cardiac hypertrophy and damage, which is mainly due to its antioxidant and anti-inflammatory potentials and requires the activation of GHS-R1a.
Collapse
MESH Headings
- Acylation
- Animals
- Anti-Inflammatory Agents/pharmacology
- Antioxidants/pharmacology
- Disease Models, Animal
- Ghrelin/analogs & derivatives
- Ghrelin/metabolism
- Ghrelin/pharmacology
- Hyperthyroidism/chemically induced
- Hyperthyroidism/metabolism
- Hypertrophy, Left Ventricular/chemically induced
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/prevention & control
- Inflammation Mediators/metabolism
- Male
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Oxidative Stress/drug effects
- Rats, Sprague-Dawley
- Reactive Nitrogen Species/metabolism
- Renin-Angiotensin System/drug effects
- Thyroxine
- Ventricular Function, Left/drug effects
- Rats
Collapse
Affiliation(s)
- Rehab Badi
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia ; and
- Department of Physiology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
10
|
Noda M, Liu J, Long J. Neuroprotective and Preventative Effects of Molecular Hydrogen. Curr Pharm Des 2021; 27:585-591. [PMID: 33076798 DOI: 10.2174/1381612826666201019103020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/12/2020] [Indexed: 11/22/2022]
Abstract
One of the beneficial effects of molecular hydrogen (H2, hydrogen gas) is neuroprotection and prevention of neurological disorders. It is important and useful if taking H2 every day can prevent or ameliorate the progression of neurodegenerative disorders, such as Parkinson's disease or Alzheimer's disease, both lacking specific therapeutic drugs. There are several mechanisms of how H2 protects neuronal damage. Anti-oxidative, anti-inflammatory, and the regulation of the endocrine system via stomach-brain connection seem to play an important role. At the cellular and tissue level, H2 appears to prevent the production of reactive oxygen species (ROS), and not only hydroxy radical (•OH) but also superoxide. In Parkinson's disease model mice, chronic intake of H2 causes the release of ghrelin from the stomach. In Alzheimer's disease model mice, sex-different neuroprotection is observed by chronic intake of H2. In female mice, declines of estrogen and estrogen receptor-β (ERβ) are prevented by H2, upregulating brain-derived neurotrophic factor (BDNF) and its receptor, tyrosine kinase receptor B (TrkB). The question of how drinking H2 upregulates the release of ghrelin or attenuates the decline of estrogen remains to be investigated and the mechanism of how H2 modulates endocrine systems and the fundamental question of what or where is the target of H2 needs to be elucidated for a better understanding of the effects of H2.
Collapse
Affiliation(s)
- Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine and Center for Translational Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine and Center for Translational Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
11
|
Potential Antioxidative, Anti-inflammatory and Immunomodulatory Effects of Ghrelin, an Endogenous Peptide from the Stomach in SARS-CoV2 Infection. Int J Pept Res Ther 2021; 27:1875-1883. [PMID: 33880115 PMCID: PMC8050636 DOI: 10.1007/s10989-021-10217-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 02/08/2023]
Abstract
The current COVID-19 pandemic is one of the most devastating events in recent history. The respiratory effects of this disease include acute respiratory distress syndrome, systemic inflammation, cytokine storm, and pulmonary fibrosis. Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor, is a peptide hormone secreted mainly by the stomach. Interestingly, ghrelin possesses promising antioxidant, anti-and inflammatory effects, making it an attractive agent to reduce the complications of the SARS-CoV-2. In addition, ghrelin exerts a wide range of immunomodulatory and anti-inflammatory effects and can mitigate the uncontrolled cytokine production responsible for acute lung injury by upregulating PPARγ and down-regulating NF-κB expression. Ghrelin has also been reported to enhance Nrf2 expression in inflammatory conditions which led to the suppression of oxidative stress. The current opinion summarizes the evidence for the possible pharmacological benefits of ghrelin in the therapeutic management of SARS-CoV-2 infection.
Collapse
|
12
|
Wang H, Dou S, Zhu J, Shao Z, Wang C, Cheng B. Regulatory effects of ghrelin on endoplasmic reticulum stress, oxidative stress, and autophagy: Therapeutic potential. Neuropeptides 2021; 85:102112. [PMID: 33333485 DOI: 10.1016/j.npep.2020.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/29/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Ghrelin is a regulatory peptide that is the endogenous ligand of the growth hormone secretagogue 1a (GHS-R1a) which belongs to the G protein-coupled receptor family. Ghrelin and GHS-R1a are widely expressed in the central and peripheral tissues and play therapeutic potential roles in the cytoprotection of many internal organs. Endoplasmic reticulum stress (ERS), oxidative stress, and autophagy dysfunction, which are involved in various diseases. In recent years, accumulating evidence has suggested that ghrelin exerts protective effects by regulating ERS, oxidative stress, and autophagy in diverse diseases. This review article summarizes information about the roles of the ghrelin system on ERS, oxidative stress, and autophagy in multiple diseases. It is suggested that ghrelin positively affects the treatment of diseases and may be considered as a therapeutic drug in many illnesses.
Collapse
Affiliation(s)
- Huiqing Wang
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Shanshan Dou
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Junge Zhu
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Ziqi Shao
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Baohua Cheng
- Neurobiology Institute, Jining Medical University, 272067 Jining, China.
| |
Collapse
|
13
|
Mathur N, Mehdi SF, Anipindi M, Aziz M, Khan SA, Kondakindi H, Lowell B, Wang P, Roth J. Ghrelin as an Anti-Sepsis Peptide: Review. Front Immunol 2021; 11:610363. [PMID: 33584688 PMCID: PMC7876230 DOI: 10.3389/fimmu.2020.610363] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Sepsis continues to produce widespread inflammation, illness, and death, prompting intensive research aimed at uncovering causes and therapies. In this article, we focus on ghrelin, an endogenous peptide with promise as a potent anti-inflammatory agent. Ghrelin was discovered, tracked, and isolated from stomach cells based on its ability to stimulate release of growth hormone. It also stimulates appetite and is shown to be anti-inflammatory in a wide range of tissues. The anti-inflammatory effects mediated by ghrelin are a result of both the stimulation of anti-inflammatory processes and an inhibition of pro-inflammatory forces. Anti-inflammatory processes are promoted in a broad range of tissues including the hypothalamus and vagus nerve as well as in a broad range of immune cells. Aged rodents have reduced levels of growth hormone (GH) and diminished immune responses; ghrelin administration boosts GH levels and immune response. The anti-inflammatory functions of ghrelin, well displayed in preclinical animal models of sepsis, are just being charted in patients, with expectations that ghrelin and growth hormone might improve outcomes in patients with sepsis.
Collapse
Affiliation(s)
- Nimisha Mathur
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Syed F. Mehdi
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Manasa Anipindi
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Sawleha A. Khan
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Hema Kondakindi
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Barbara Lowell
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Ping Wang
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Jesse Roth
- Laboratory of Diabetes, Obesity, and Other Metabolic Disorders, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| |
Collapse
|
14
|
Reich N, Hölscher C. Acylated Ghrelin as a Multi-Targeted Therapy for Alzheimer's and Parkinson's Disease. Front Neurosci 2020; 14:614828. [PMID: 33381011 PMCID: PMC7767977 DOI: 10.3389/fnins.2020.614828] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Much thought has been given to the impact of Amyloid Beta, Tau and Alpha-Synuclein in the development of Alzheimer's disease (AD) and Parkinson's disease (PD), yet the clinical failures of the recent decades indicate that there are further pathological mechanisms at work. Indeed, besides amyloids, AD and PD are characterized by the culminative interplay of oxidative stress, mitochondrial dysfunction and hyperfission, defective autophagy and mitophagy, systemic inflammation, BBB and vascular damage, demyelination, cerebral insulin resistance, the loss of dopamine production in PD, impaired neurogenesis and, of course, widespread axonal, synaptic and neuronal degeneration that leads to cognitive and motor impediments. Interestingly, the acylated form of the hormone ghrelin has shown the potential to ameliorate the latter pathologic changes, although some studies indicate a few complications that need to be considered in the long-term administration of the hormone. As such, this review will illustrate the wide-ranging neuroprotective properties of acylated ghrelin and critically evaluate the hormone's therapeutic benefits for the treatment of AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical & Life Sciences Division, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, A Second Hospital, Shanxi Medical University, Taiyuan, China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
15
|
Ferrer B, Prince LM, Tinkov AA, Santamaria A, Bowman AB, Aschner M. Chronic exposure to methylmercury disrupts ghrelin actions in C57BL/6J mice. Food Chem Toxicol 2020; 147:111918. [PMID: 33301842 DOI: 10.1016/j.fct.2020.111918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 01/06/2023]
Abstract
Methylmercury (MeHg) is a neurotoxic pollutant widely present in the environment. Initial symptoms of MeHg may include loss of body weight. However, the mechanisms by which MeHg induces body weight changes have yet to be fully elucidated. Body weight is regulated by multiple mechanisms. Whereas multiple peripheral peptides lead to food intake cessation, ghrelin is the only recognized peripheral hormone that stimulates food intake. It exerts its action on Neuropeptide Y/Agouti-related peptide neurons in the hypothalamus. To test if MeHg affects ghrelin signaling C57BL/6J mice (males and females) were exposed to 5 ppm MeHg via drinking water during a month. On days 15 and 30 of MeHg exposure ghrelin was administered intraperitoneally and changes in body weight and food intake were recorded. In addition, changes in ghrelin-induced signaling pathways in hypothalamus were also analyzed. Here, we show that in males, MeHg enhanced ghrelin-induced body weight gain by activating the AMP-activated Kinase (AMPK)/Uncoupled protein 2 (UCP2) signaling pathway. In contrast, in females, MeHg inhibited ghrelin-induced mTOR signaling activation and decreased Npy mRNA expression, thus mitigating the ghrelin-induced weight gain. Combined, our novel results demonstrate, for the first time, that MeHg disrupts the physiological functions of ghrelin differently in males and females.
Collapse
Affiliation(s)
- Beatriz Ferrer
- Department of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA.
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia.
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico.
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Michael Aschner
- Department of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA; IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia.
| |
Collapse
|
16
|
Breithaupt L, Chunga-Iturry N, Lyall AE, Cetin-Karayumak S, Becker KR, Thomas JJ, Slattery M, Makris N, Plessow F, Pasternak O, Holsen LM, Kubicki M, Misra M, Lawson EA, Eddy KT. Developmental stage-dependent relationships between ghrelin levels and hippocampal white matter connections in low-weight anorexia nervosa and atypical anorexia nervosa. Psychoneuroendocrinology 2020; 119:104722. [PMID: 32512249 PMCID: PMC8629489 DOI: 10.1016/j.psyneuen.2020.104722] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/29/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Disruptions in homeostatic and hedonic food motivation are proposed to underlie anorexia nervosa (AN) and atypical AN, restrictive eating disorders which commonly onset in puberty. Ghrelin, a neuroprotective hormone that drives hedonic eating is increased in AN and is expressed in the hippocampus. White matter (WM) undergoes significant change during puberty in regions involved in food motivation, particularly WM tracts connected with the hippocampus. The association between ghrelin and WM region of interest (ROI) with hippocampal connections in restrictive eating disorders, particularly in adolescence during key neurodevelopmental growth, is unknown. METHODS We evaluated fasting plasma ghrelin and WM microstructure (measured by free-water corrected fractional anisotropy (FA-t)) in WM ROIs with hippocampal connections - the fornix and the hippocampal portion of the cingulum - in 56 adolescent females (age range: 11.9 - 22.1 y; mean: 19.0 y) with low-weight eating disorders including AN and atypical AN (N = 36) and healthy controls (N = 20). RESULTS FA-t in the fornix or hippocampal portion of the fornix did not differ between groups. Ghrelin was higher in AN/atypical AN vs. HC and was positively correlated with puberty stage in the AN/atypical AN group, but not the HC group. The correlation between ghrelin and FA-t in the fornix was significantly different in females with AN/atypical AN compared to controls. In AN/atypical AN, pubertal stage moderated the relation between fasting plasma ghrelin and FA-t in the fornix: higher fasting ghrelin was associated with lower FA-t in the fornix in late-post-puberty, but was not associated with FA-t in the early to mid stages of puberty. CONCLUSIONS In post-pubertal females with low-weight AN/atypical AN, higher levels of ghrelin are associated with lower FA-t in the fornix. This relationship is not evident in the early to mid stages of puberty in AN/atypical AN or in HC, and may reflect a lack of possible neuroprotective effects of ghrelin in late-post puberty only. Understanding the effects of ghrelin on WM microstructure longitudinally and following recovery from AN/Atypical AN and how this differs across pubertal stages will be an important next step. These findings could ultimately inform treatment staging and aid in diagnosis and detection of AN/atypical AN.
Collapse
Affiliation(s)
- Lauren Breithaupt
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Natalia Chunga-Iturry
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA
| | - Amanda E Lyall
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA
| | - Suheyla Cetin-Karayumak
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA
| | - Kendra R Becker
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jennifer J Thomas
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Meghan Slattery
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Nikos Makris
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA; Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States of America; Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Franziska Plessow
- Harvard Medical School, Boston, MA, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Ofer Pasternak
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA; Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Laura M Holsen
- Harvard Medical School, Boston, MA, USA; Division of Women's Health, Brigham and Women's Hospital, Boston, MA, USA
| | - Marek Kubicki
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA; Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States of America; Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Madhusmita Misra
- Harvard Medical School, Boston, MA, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA; Division of Pediatric Endocrinology, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth A Lawson
- Harvard Medical School, Boston, MA, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Kamryn T Eddy
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Meloni BP, Mastaglia FL, Knuckey NW. Cationic Arginine-Rich Peptides (CARPs): A Novel Class of Neuroprotective Agents With a Multimodal Mechanism of Action. Front Neurol 2020; 11:108. [PMID: 32158425 PMCID: PMC7052017 DOI: 10.3389/fneur.2020.00108] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
There are virtually no clinically available neuroprotective drugs for the treatment of acute and chronic neurological disorders, hence there is an urgent need for the development of new neuroprotective molecules. Cationic arginine-rich peptides (CARPs) are an expanding and relatively novel class of compounds, which possess intrinsic neuroprotective properties. Intriguingly, CARPs possess a combination of biological properties unprecedented for a neuroprotective agent including the ability to traverse cell membranes and enter the CNS, antagonize calcium influx, target mitochondria, stabilize proteins, inhibit proteolytic enzymes, induce pro-survival signaling, scavenge toxic molecules, and reduce oxidative stress as well as, having a range of anti-inflammatory, analgesic, anti-microbial, and anti-cancer actions. CARPs have also been used as carrier molecules for the delivery of other putative neuroprotective agents across the blood-brain barrier and blood-spinal cord barrier. However, there is increasing evidence that the neuroprotective efficacy of many, if not all these other agents delivered using a cationic arginine-rich cell-penetrating peptide (CCPPs) carrier (e.g., TAT) may actually be mediated largely by the properties of the carrier molecule, with overall efficacy further enhanced according to the amino acid composition of the cargo peptide, in particular its arginine content. Therefore, in reviewing the neuroprotective mechanisms of action of CARPs we also consider studies using CCPPs fused to a putative neuroprotective peptide. We review the history of CARPs in neuroprotection and discuss in detail the intrinsic biological properties that may contribute to their cytoprotective effects and their usefulness as a broad-acting class of neuroprotective drugs.
Collapse
Affiliation(s)
- Bruno P Meloni
- Department of Neurosurgery, QEII Medical Centre, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Frank L Mastaglia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Neville W Knuckey
- Department of Neurosurgery, QEII Medical Centre, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
18
|
Zhang HB, Tu XK, Song SW, Liang RS, Shi SS. Baicalin Reduces Early Brain Injury after Subarachnoid Hemorrhage in Rats. Chin J Integr Med 2020; 26:510-518. [DOI: 10.1007/s11655-020-3183-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2018] [Indexed: 10/25/2022]
|
19
|
Cheng Y, Chen B, Xie W, Chen Z, Yang G, Cai Y, Shang H, Zhao W. Ghrelin attenuates secondary brain injury following intracerebral hemorrhage by inhibiting NLRP3 inflammasome activation and promoting Nrf2/ARE signaling pathway in mice. Int Immunopharmacol 2020; 79:106180. [PMID: 31926478 DOI: 10.1016/j.intimp.2019.106180] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/16/2019] [Accepted: 12/31/2019] [Indexed: 02/08/2023]
Abstract
Ghrelin, a brain-gut peptide, has been proven to exert neuroprotection in different kinds of neurological diseases; however, its role and the potential molecular mechanisms in secondary brain injury (SBI) after intracerebral hemorrhage (ICH) are still unknown. In this study, we investigate whether treatment with ghrelin may attenuate SBI in a murine ICH model, and if so, whether the neuroprotective effects are due to the inhibition of nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome activation and promotion of nuclear factor-E2-related factor 2 (Nrf2)/antioxidative response element (ARE) signaling pathway. Stereotactically intrastriatal infusion of autologous blood was performed to mimic ICH. Ghrelin was given intraperitoneally immediately following ICH and again 1 h later. Results showed that ghrelin attenuated neurobehavioral deficits, brain edema, hematoma volume, and perihematomal cell death post-ICH. Ghrelin inhibited the NLRP3 inflammasome activation and subsequently suppressed the neuroinflammatory response as evidenced by reduced microglia activation, neutrophil infiltration, and pro-inflammatory mediators release after ICH. Additionally, ghrelin alleviated ICH-induced oxidative stress according to the chemiluminescence of luminol and lucigenin, malondialdehyde (MDA) content, and total superoxide dismutase (SOD) activity assays. These changes were accompanied by upregulation of Nrf2 expression, Nrf2 nuclear accumulation, and enhanced Nrf2 DNA binding activity, as well as by increased expressions of Nrf2 downstream target antioxidative genes, including NAD(P)H quinine oxidoreductase-1 (NQO1), glutathione cysteine ligase regulatory subunit (GCLC), and glutathione cysteine ligase modulatory subunit (GCLM). Together, our data suggested that ghrelin protected against ICH-induced SBI by inhibiting NLRP3 inflammasome activation and promoting Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Yijun Cheng
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Bin Chen
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Wanqun Xie
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Zhenghong Chen
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Guoyuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, PR China; Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yu Cai
- Department of Neurosurgery, North Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hanbing Shang
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Weiguo Zhao
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
20
|
Koyuncuoğlu T, Arabacı Tamer S, Erzik C, Karagöz A, Akakın D, Yüksel M, Yeğen BÇ. Oestrogen receptor ERα and ERβ agonists ameliorate oxidative brain injury and improve memory dysfunction in rats with an epileptic seizure. Exp Physiol 2019; 104:1911-1928. [PMID: 31608530 DOI: 10.1113/ep087986] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022]
Abstract
NEW FINDINGS What is the central question of this study? Could different hormonally active substances, including oestrogen receptor (ER) agonists, protect against oxidative brain damage and memory impairment induced by a single epileptic seizure in rats? If so, which signalling mechanisms are involved in their anti-inflammatory effects? What is the main finding and its importance? Chronic administration of oestrogen, progesterone, ER modulators/agonists or blockade of testosterone exhibited anti-inflammatory and antioxidant actions on single seizure-induced neuronal injury, while ER agonists additionally improved memory function and up-regulated CREB signalling and hippocampal GABA(A)α1 receptor density, suggesting that ERα or ERβ receptor activation may be beneficial in protecting against seizure-related oxidative brain injury and cognitive dysfunction. ABSTRACT The susceptibility to epileptic seizures is dependent on sex as well as fluctuations in oestrogen levels, while exogenous oestrogen was shown to have no effect or to facilitate or to inhibit seizure activity. Oestrogen receptors (ERs) mediate antioxidant and anti-inflammatory actions in several inflammatory models, but the involvement of ERs in seizure-induced neuronal injury has not been evaluated previously. In order to assess the effects of resveratrol, progesterone, oestradiol (E2), an anti-testosterone (cyproterone acetate; CPA), a selective ER modulator (tamoxifen; TMX) and ERα/ERβ agonists (propyl pyrazole triol (PPT), diarylpropionitrile (DPN)) on oxidative brain damage and memory impairment due to epileptic seizure, male Wistar rats (n = 120) received one of the treatment choices either in drinking water or intraperitoneally for 31 days, and epileptic seizure was induced on the 28th day by injection of a single-dose of pentylenetetrazole (45 mg kg-1 ). The results demonstrate that chronic pretreatment with resveratrol, progesterone, E2, CPA or TMX suppressed most of the inflammatory parameters indicative of oxidative neuronal injury, while treatment with the ER agonists DPN or PPT were found to be even more effective in limiting the oxidative damage. Treatment with DPN resulted in the up-regulation of cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) expression, while PPT up-regulated expression of CREB without affecting BDNF levels. Moreover, both ER agonists provided protection against seizure-induced memory loss with a concomitant increase in hippocampal GABA(A)α1-positive cells. In conclusion, ER agonists, and more specifically ERβ agonist, appear to provide maximum protection against seizure-induced oxidative brain injury and associated memory dysfunction by up-regulating the expression of CREB, BDNF and GABA(A)α1 receptors.
Collapse
Affiliation(s)
- Türkan Koyuncuoğlu
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Sevil Arabacı Tamer
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Can Erzik
- Department of Medical Biology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ayça Karagöz
- Department of Histology & Embryology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Dilek Akakın
- Department of Histology & Embryology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Meral Yüksel
- Department of Medical Laboratory, Vocational School of Health-Related Professions, Marmara University, Istanbul, Turkey
| | - Berrak Ç Yeğen
- Department of Physiology, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
21
|
Tyurina YY, St Croix CM, Watkins SC, Watson AM, Epperly MW, Anthonymuthu TS, Kisin ER, Vlasova II, Krysko O, Krysko DV, Kapralov AA, Dar HH, Tyurin VA, Amoscato AA, Popova EN, Bolevich SB, Timashev PS, Kellum JA, Wenzel SE, Mallampalli RK, Greenberger JS, Bayir H, Shvedova AA, Kagan VE. Redox (phospho)lipidomics of signaling in inflammation and programmed cell death. J Leukoc Biol 2019; 106:57-81. [PMID: 31071242 PMCID: PMC6626990 DOI: 10.1002/jlb.3mir0119-004rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 04/12/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
In addition to the known prominent role of polyunsaturated (phospho)lipids as structural blocks of biomembranes, there is an emerging understanding of another important function of these molecules as a highly diversified signaling language utilized for intra- and extracellular communications. Technological developments in high-resolution mass spectrometry facilitated the development of a new branch of metabolomics, redox lipidomics. Analysis of lipid peroxidation reactions has already identified specific enzymatic mechanisms responsible for the biosynthesis of several unique signals in response to inflammation and regulated cell death programs. Obtaining comprehensive information about millions of signals encoded by oxidized phospholipids, represented by thousands of interactive reactions and pleiotropic (patho)physiological effects, is a daunting task. However, there is still reasonable hope that significant discoveries, of at least some of the important contributors to the overall overwhelmingly complex network of interactions triggered by inflammation, will lead to the discovery of new small molecule regulators and therapeutic modalities. For example, suppression of the production of AA-derived pro-inflammatory mediators, HXA3 and LTB4, by an iPLA2 γ inhibitor, R-BEL, mitigated injury associated with the activation of pro-inflammatory processes in animals exposed to whole-body irradiation. Further, technological developments promise to make redox lipidomics a powerful approach in the arsenal of diagnostic and therapeutic instruments for personalized medicine of inflammatory diseases and conditions.
Collapse
Affiliation(s)
- Yulia Y Tyurina
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Claudette M St Croix
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alan M Watson
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tamil S Anthonymuthu
- Department of Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elena R Kisin
- Exposure Assessment Branch, NIOSH/CDC, Morgantown, West Virginia, USA
| | - Irina I Vlasova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Moscow, Russia
- Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Olga Krysko
- Upper Airways Research Laboratory, Department of Head and Skin, Ghent University, and Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, and Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Alexandr A Kapralov
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Haider H Dar
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vladimir A Tyurin
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew A Amoscato
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elena N Popova
- Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Sergey B Bolevich
- Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Peter S Timashev
- Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Moscow, Russia
| | - John A Kellum
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hulya Bayir
- Department of Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anna A Shvedova
- Exposure Assessment Branch, NIOSH/CDC, Morgantown, West Virginia, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Laboratory of Navigational Redox Lipidomics, IM Sechenov Moscow State Medical University, Moscow, Russia
| |
Collapse
|
22
|
ÇİMEN S, TAŞDEMİR C, VARDI N, ATEŞ B, TAŞDEMİR S, ÖZAYDOĞDU ÇİMEN A. Protective effects of ghrelin on kidney tissue in rats with partial ureteral obstruction. Turk J Med Sci 2019; 49:696-702. [PMID: 30997983 PMCID: PMC7018211 DOI: 10.3906/sag-1802-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background/aim The aim was to investigate the protective and therapeutic effects of ghrelin, which has antioxidant and antiinflammatory activity, on preventing kidney damage that occurs by induced partial ureteral obstruction in rats Materials and methods Twenty-eight adult male rats were included in the study, and the rats were divided into 4 groups. After the laparotomy operation on the sham group, the ureter was identified in the retroperitoneal area and was duly sutured (n = 7). Ghrelin was administered for seven days intraperitoneally, and after the nephrectomy performed on the 15th day, the rats were sacrificed (n = 7). A partial ureteral obstruction was performed after the laparotomy on the PUO group. The rats were sacrificed after the nephrectomy operation performed on the 15th day (n = 7). A partial ureteral obstruction was formed after the laparotomy followed by seven days of waiting in the PUO + ghrelin group. Ghrelin was given in the dose of 10 ng/kg per day intraperitoneally for the next 7 days, and the rats were sacrificed after the nephrectomy operation performed on the 15th day (n = 7). All groups were evaluated for histological damage and catalase, superoxide dismutase, total glutathione, malondialdehyde, and myeloperoxidase levels were measured in the same tissues Results When the 2nd group and the sham group were compared histologically, it was observed that the damage had increased by a statistically significant level in the partial ureteral obstruction group (P = 0.001). When the group which was ghrelin-treated after the partial ureteral obstruction was compared to the group with just partial ureteral obstruction, the histopathological changes were found to decrease significantly in that group (P = 0.001). While the statistical significance of the levels of CAT, GSH, and MPO enzymes was detected among biochemical changes in the 2nd group when compared to the sham group (P < 0.01), the 3rd group showed a statistically significant difference in the levels of SOD and GSH enzymes compared to the 4th group (P < 0.05). Conclusion Ghrelin administration to rats after the formation of an experimental partial unilateral ureteral obstruction reduces tissue damage due to ghrelin’s antiinflammatory and antioxidant effects. Ghrelin administration may prevent tissue damage biochemically and histopathologically in obstructive uropathy cases
Collapse
Affiliation(s)
- Serhan ÇİMEN
- Department of Urology, Malatya Training and Research Hospital, MalatyaTurkey
- * To whom correspondence should be addressed. E-mail:
| | - Cemal TAŞDEMİR
- Department of Urology, Turgut Özal Medical Center, İnönü University, MalatyaTurkey
| | - Nigar VARDI
- Department of Histology, Turgut Özal Medical Center, İnönü University, MalatyaTurkey
| | - Burhan ATEŞ
- Department of Chemistry, Faculty of Science, İnönü University, MalatyaTurkey
| | - Seda TAŞDEMİR
- Department of Pharmacology, Turgut Özal Medical Center, İnönü University, MalatyaTurkey
| | | |
Collapse
|
23
|
Boyacı MG, Rakip U, Aslan A, Koca HB, Aslan E, Korkmaz S, Yıldızhan S. Effects of 2-Aminoethyl Diphenylborinate, a Modulator of Transient Receptor Potential and Orai Channels in Subarachnoid Hemorrhage: An Experimental Study. World Neurosurg 2019; 127:e376-e388. [PMID: 30905651 DOI: 10.1016/j.wneu.2019.03.129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cerebral vasospasm remains a serious problem affecting morbidity and mortality in patients with subarachnoid hemorrhage (SAH) during neurosurgery. We aimed to demonstrate the role of the transient receptor potential channel and other channels for Ca2+ in the etiology of cerebral vasospasm using 2-aminoethyl diphenylborinate (2-APB) and the effective dose range of an unstudied pharmacological agent, which can limit vasospasm. METHODS We performed an experimental study using 32 Sprague-Dawley rats divided into 4 groups: sham group (n = 8), SAH group (n = 8), 2-APB group (SAH rats intraperitoneally administered with 0.5 mg/kg 2-APB; n = 8), and 2-APB-2 group (SAH rats intraperitoneally administered with 2 mg/kg 2-APB; n = 8). The rats were sacrificed after 24 hours, and superoxide dismutase, glutathione peroxidase, malondialdehyde, tumor necrosis factor-α, and interleukin-1β in the brain tissue and serum were measured. The histopathological investigation of brain tissue included measurement of the luminal diameter and wall thickness of the basilar artery (BA), and apoptotic cells in the hippocampus were counted after caspase staining. RESULTS Autologous arterial blood injection into the cisterna magna caused vasospasm in rats. 2-APB treatment increased the BA wall thickness and reduced the BA lumen diameter, inducing significant vascular changes. 2-APB also alleviated cell apoptosis at 24 hours after SAH. CONCLUSION In experimental SAH in rats, 2-APB treatment increased the BA wall thickness and reduced the BA lumen diameter, inducing significant vascular changes. 2-APB also alleviated cell apoptosis at 24 hours after SAH.
Collapse
Affiliation(s)
- Mehmet Gazi Boyacı
- Department of Neurosurgery, Afyonkarahisar Health Sciences University Faculty of Medicine, Afyonkarahisar, Turkey.
| | - Usame Rakip
- Department of Neurosurgery, TCSB Niğde Ömer Halisdemir University Hospital, Niğde, Turkey
| | - Adem Aslan
- Department of Neurosurgery, Afyonkarahisar Health Sciences University Faculty of Medicine, Afyonkarahisar, Turkey
| | - Halit Buğra Koca
- Department of Biochemistry, Afyonkarahisar Health Sciences University Faculty of Medicine, Afyonkarahisar, Turkey
| | - Esra Aslan
- Department of Histology and Embryology, Afyonkarahisar Health Sciences University Faculty of Medicine, Afyonkarahisar, Turkey
| | - Serhat Korkmaz
- Department of Neurosurgery, Afyonkarahisar Health Sciences University Faculty of Medicine, Afyonkarahisar, Turkey
| | - Serhat Yıldızhan
- Department of Neurosurgery, Afyonkarahisar Health Sciences University Faculty of Medicine, Afyonkarahisar, Turkey
| |
Collapse
|
24
|
Toklu HZ, Yang Z, Ersahin M, Wang KKW. Neurological Exam in Rats Following Stroke and Traumatic Brain Injury. Methods Mol Biol 2019; 2011:371-381. [PMID: 31273710 DOI: 10.1007/978-1-4939-9554-7_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Using the appropriate model for testing neurological symptoms in rats is essential for the assessment of functional outcome. A number of tests have been developed to quantify the severity of neurological deficits. These tests should meet criteria such as validity, specificity, sensitivity, and utility. Although analysis of motor function shows homology in primates and rodents, the total neurological exam scores may not always reflect the clinical outcome. Therefore, the selection of the appropriate tests has critical importance when evaluating therapeutic strategies. This chapter describes Toklu's modified neurological exam score method which can be used practically to assess neurological symptoms following traumatic brain injury (TBI) and stroke. The method is a combination of balance, muscle strength, coordination, and reflex.
Collapse
Affiliation(s)
- Hale Z Toklu
- University of Central Florida College of Medicine, Department of Clinical Sciences, Gainesville, FL, USA.
- HCA North Florida Division, Graduate Medical Education, Tallahassee, FL, USA.
| | - Zhiui Yang
- University of Florida, Department of Emergency Medicine, Gainesville, FL, USA
| | - Mehmet Ersahin
- Istanbul Medeniyet University, Department of Neurosurgery, Istanbul, Turkey
| | - Kevin K W Wang
- University of Florida, Department of Emergency Medicine, Gainesville, FL, USA
| |
Collapse
|
25
|
Aykac A, Ozbeyli D, Uncu M, Ertaş B, Kılınc O, Şen A, Orun O, Sener G. Evaluation of the protective effect of Myrtus communis in scopolamine-induced Alzheimer model through cholinergic receptors. Gene 2018; 689:194-201. [PMID: 30553998 DOI: 10.1016/j.gene.2018.12.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/15/2018] [Accepted: 12/07/2018] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder causing common health problem with increasing age. Evidences show that the key symptoms of AD are mainly caused by cholinergic system dysfunction which has a role in cognitive disorders. Cholinergic pathways especially muscarinic receptors like M1 subtype also have a major role in learning, memory, cognitive functions and emotional state. There is no available permanent treatment currently to cure AD or to change its progression. This study was designed to investigate the factors that play important role in pathogenesis of AD and to compare the effects of Galantamine treatment with effects of Myrtus communis treatment. The expression level of M1, ACh, BDNF; AChE activity, GSH level, MDA and MPO activity and AChE gene expression were investigated in scopolamine-induced rat model. Results showed that, administration of MC significantly improves the SCOP-induced reduction of latency and object recognition time; increasing BDNF, M1 and ACh receptor expression levels in the different brain regions. Additionally, MC showed an increased in AChE by enhancing GSH activity and reducing MDA level and MPO activity. In conclusion MC considered as a possible novel therapeutic approach that can be a valuable alternative way in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Asli Aykac
- Department of Biophysics, Faculty of Medicine, Near East University, Nicosia, Cyprus.
| | - Dilek Ozbeyli
- Vocational School of Health Services, Marmara University, Istanbul, Turkey
| | - Murat Uncu
- Department of Clinical Biochemistry, Dr. Burhan Nalbantoglu State Hospital, Nicosia, Cyprus
| | - Büşra Ertaş
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Olca Kılınc
- Department of Biophysics, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Ali Şen
- Department of Pharmacognosy, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Oya Orun
- Department of Biophysics, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Goksel Sener
- Department of Pharmacology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| |
Collapse
|
26
|
Lyra Junior HF, de Lucca Schiavon L, Rodrigues IK, Couto Vieira DS, de Paula Martins R, Turnes BL, Latini AS, D'Acâmpora AJ. Effects of Ghrelin on the Oxidative Stress and Healing of the Colonic Anastomosis in Rats. J Surg Res 2018; 234:167-177. [PMID: 30527470 DOI: 10.1016/j.jss.2018.09.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Anastomotic leakage is the deadliest complication of colonic procedures. Ghrelin is an orexigenic hormone with potent actions on growth hormone release and functions in the processes of growth, tissue inflammation, repair, and oxidative stress. We evaluated the hypothesis that the exogenous administration of ghrelin causes beneficial effects on the healing of colonic anastomosis. MATERIALS AND METHODS Sixty-four male Wistar rats were randomly assigned to eight subgroups receiving postoperative intraperitoneal administration of ghrelin (23 μg/kg/d) or saline after a colonic anastomosis. The anastomotic tissue was evaluated on the third, seventh, and 14th postoperative days. Anastomotic bursting pressure, histological parameters, hydroxyproline content, and tissue oxidative stress markers were compared. RESULTS There was a significant increase in the mean anastomotic bursting pressure in the ghrelin subgroup on the seventh postoperative day (P = 0.035). Histological evaluation demonstrated a significant difference in the neutrophilic infiltrate (P = 0.035) on the third and 14th d and in apoptosis (P = 0.004), granulation tissue (P = 0.011) and peritoneal inflammation (P = 0.014) on the 14th postoperative day. There was a statistically significant increase in the hydroxyproline content in the ghrelin subgroup on the 14th postoperative day (P = 0.043). There were significant differences in the nitrite tissue levels (P = 0.021) on day 3 and in reactive oxygen species (P = 0.012) on day 14. CONCLUSIONS The administration of ghrelin had beneficial anti-inflammatory and antioxidant effects, increasing the resistance of the anastomosis and the hydroxyproline tissue content in the postoperative period.
Collapse
Affiliation(s)
| | - Leonardo de Lucca Schiavon
- Department of Internal Medicine, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Igor Kunze Rodrigues
- Department of Surgery, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | | | - Roberta de Paula Martins
- Department of Biochemistry, Bioenergetics and Oxidative Stress Laboratory, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Bruna Lenfers Turnes
- Department of Biochemistry, Bioenergetics and Oxidative Stress Laboratory, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Alexandra Susana Latini
- Department of Biochemistry, Bioenergetics and Oxidative Stress Laboratory, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Armando José D'Acâmpora
- Department of Surgery, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| |
Collapse
|
27
|
Anthonymuthu TS, Kenny EM, Lamade AM, Kagan VE, Bayır H. Oxidized phospholipid signaling in traumatic brain injury. Free Radic Biol Med 2018; 124:493-503. [PMID: 29964171 PMCID: PMC6098726 DOI: 10.1016/j.freeradbiomed.2018.06.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/21/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022]
Abstract
Oxidative stress is a major contributor to secondary injury signaling cascades following traumatic brain injury (TBI). The role of lipid peroxidation in the pathophysiology of a traumatic insult to neural tissue is increasingly recognized. As the methods to quantify lipid peroxidation have gradually improved, so has the understanding of mechanistic details of lipid peroxidation and related signaling events in the injury pathogenesis. While free-radical mediated, non-enzymatic lipid peroxidation has long been studied, recent advances in redox lipidomics have demonstrated the significant contribution of enzymatic lipid peroxidation to TBI pathogenesis. Complex interactions between inflammation, phospholipid peroxidation, and hydrolysis define the engagement of different cell death programs and the severity of injury and outcome. This review focuses on enzymatic phospholipid peroxidation after TBI, including the mechanism of production, signaling roles in secondary injury pathology, and temporal course of production with respect to inflammatory response. In light of the newly identified phospholipid oxidation mechanisms, we also discuss possible therapeutic targets to improve neurocognitive outcome after TBI. Finally, we discuss current limitations in identifying oxidized phospholipids and possible methodologic improvements that can offer a deeper insight into the region-specific distribution and subcellular localization of phospholipid oxidation after TBI.
Collapse
Affiliation(s)
- Tamil S Anthonymuthu
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Elizabeth M Kenny
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Andrew M Lamade
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, United States; Laboratory of Navigational Redox Lipidomics in Biomedicine, Department of Human Pathology, IM Sechenov First Moscow State Medical University, Russian Federation
| | - Hülya Bayır
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, United States; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, 15224, United States.
| |
Collapse
|
28
|
Administration of ghrelin associated with decreased expression of matrix metalloproteinase-9 following normobaric systemic hypoxia in the brain. Endocr Regul 2018; 52:152-158. [PMID: 31517605 DOI: 10.2478/enr-2018-0019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE According to our previous studies, ghrelin protects blood brain barrier (BBB) integrity and it attenuates hypoxia-induced brain edema in the hypoxic conditions. However, the underlying mechanisms remain poorly understood. Several studies suggest a role for matrix metal-loproteinase-9 (MMP9) in the BBB disruption and cerebral edema formation. The present study was conducted to determine the effect of ghrelin on MMP9 protein expression in the model of acute and chronic systemic hypoxia. METHODS Adult male Wistar rats were divided into acute or chronic controls, acute or chronic hypoxia and ghrelin-treated acute or chronic hypoxia groups. The hypoxic groups were kept in the hypoxic chamber (10-11% O2) for two (acute) or ten days (chronic). Effect of ghrelin on MMP9 protein expression was assessed using immunoblotting. RESULTS Our results showed that acute and chronic systemic hypoxia increased the MMP9 protein expression in the brain (p<0.001). Treatment with ghrelin significantly attenuated this expression in the cerebral hypoxia (p<0.05). CONCLUSION Our results demonstrate that the neuroprotective effects of ghrelin may be mediated, in part, by decreasing in MMP9 production in the hypoxic brain.
Collapse
|
29
|
Eraslan N, Elgin U, Şen E, Kilic A, Yilmazbas P. Comparison of total/active ghrelin levels in primary open angle glaucoma, pseudoexfoliation glaucoma and pseudoexfoliation syndrome. Int J Ophthalmol 2018; 11:823-827. [PMID: 29862183 DOI: 10.18240/ijo.2018.05.18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/06/2018] [Indexed: 01/06/2023] Open
Abstract
AIM To investigate the levels of ghrelin (Gh), acylated ghrelin (AGh) and AGh/Gh ratio in the humor aqueous (HA) of cases with pseudoexfoliation syndrome (PXS), pseudoexfoliation glaucoma (PXG), primary open angle glaucoma (POAG) and to compare these with control subjects. METHODS A prospective examination was made of the total Gh, and AGh levels in HA of 67 patients undergoing cataract surgery. Patients were divided into 4 groups. HA samples were aspirated at the beginning of the surgery, stored at -70°C. Gh and AGh quantification was performed with ELISA kits and the AGh/total-Gh ratios were calculated. ANOVA, Kruskal-Wallis, Chi-square and post-hoc tests were used for statistical analysis. RESULTS Total Gh levels in HA were 189.2±45.6 pg/mL in the control group, 199.2±32.9 pg/mL in PXS, 180.6±20.9 pg/mL in PXG and 176.8±21.4 pg/mL in POAG groups (P>0.05). AGh levels in HA were 23.09±5.01 pg/mL in the control group, 24.13±5.22 pg/mL in PXS, 22.29±1.55 pg/mL in PXG and 19.69±2.93 pg/mL in POAG groups (P>0.05). The ratio of AGh/Gh was 10.3%±2.34% in the control group, 13.03%±2.58% in PXS, 12.3%±1.54% in PXG and 11.79%±1.41% in POAG groups (P=0.044). The difference between the PXS and control groups was significant (P=0.03). CONCLUSION In spite of statistically insignificant results, the HA total Gh levels were lower than those of the control subjects but not parallel with the AGh levels in glaucoma patients. The relative increase in the AGh/Gh ratio in glaucoma cases supports the view that proportional increases of AGh might play a role in the pathogenesis of glaucoma.
Collapse
Affiliation(s)
- Numan Eraslan
- Ulucanlar Eye Research Hospital, Ankara 06250, Turkey
| | - Ufuk Elgin
- Ulucanlar Eye Research Hospital, Ankara 06250, Turkey
| | - Emine Şen
- Ulucanlar Eye Research Hospital, Ankara 06250, Turkey
| | - Aytul Kilic
- Department of Biochemistry, Ankara Training and Research Hospital, Ankara 06250, Turkey
| | | |
Collapse
|
30
|
Toklu HZ, Yang Z, Oktay S, Sakarya Y, Kirichenko N, Matheny MK, Muller-Delp J, Strang K, Scarpace PJ, Wang KK, Tümer N. Overpressure blast injury-induced oxidative stress and neuroinflammation response in rat frontal cortex and cerebellum. Behav Brain Res 2018; 340:14-22. [DOI: 10.1016/j.bbr.2017.04.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 12/12/2022]
|
31
|
Chung H, Choi J, Park S. Ghrelin protects adult rat hippocampal neural stem cells from excessive autophagy during oxygen-glucose deprivation. Endocr J 2018; 65:63-73. [PMID: 29057768 DOI: 10.1507/endocrj.ej17-0281] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Ghrelin functions as a neuroprotective agent and saves neurons from various insults include ischemic injury. However, it remains to be elucidated whether ghrelin protects neuronal cells against ischemic injury-induced excessive autophagy. Autophagy is required for the maintenance of neural stem cell homeostasis. However, regarding autophagic cell death, it is commonly assumed that excessive autophagy leads to self-elimination of mammalian cells. The purpose of this study was to investigate the potential neuroprotection effects of ghrelin from excessive autophagy in adult rat hippocampal neural stem cells (NSCs). Oxygen-Glucose Deprivation (OGD) strongly induces autophagy in adult rat hippocampal NSCs. Ghrelin treatment inhibited OGD-induced cell death of adult rat hippocampal NSCs assessed by cell-counting-kit-8 assay. Ghrelin also suppressed OGD-induced excessive autophagy activity. The protective effect of ghrelin was accompanied by an increased expression levels of Bcl-2, p-62 and decreased expression level of LC3-II, Beclin-1 by Western blot. Furthermore, ghrelin reduced autophagosome formation and number of GFP-LC3 transfected puncta. In conclusion, our data suggest that ghrelin protects adult rat hippocampal NSCs from excessive autophagy in experimental stroke (oxygen-glucose deprivation) model. Regulating autophagic activity may be a potential optimizing target for promoting adult rat hippocampal NSCs based therapy for stroke.
Collapse
Affiliation(s)
- Hyunju Chung
- Department of Core Research Laboratory, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Junghyun Choi
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Seungjoon Park
- Department of Pharmacology and Medical Research Center for Bioreaction to ROS and Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
32
|
Ghrelin Protects Human Lens Epithelial Cells against Oxidative Stress-Induced Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1910450. [PMID: 29129986 PMCID: PMC5654336 DOI: 10.1155/2017/1910450] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/18/2017] [Accepted: 04/23/2017] [Indexed: 12/13/2022]
Abstract
Oxidative stress has been recognized as an important mediator in the pathogenesis of age-related cataracts; using antioxidant supplements is one plausible strategy to protect the antioxidative defense system against oxidative stress. Ghrelin administration is expected to reduce ROS, preventing the onset of different diseases. The role of ghrelin, if any, in protecting against oxidative stress in HLECs has never been examined. In the present study, we investigated the effects of ghrelin against H2O2-induced oxidative stress and the associated molecular mechanisms in HLECs and rat lenses. The results showed that pretreatment with ghrelin reduced H2O2-induced cellular apoptosis and ROS accumulation, increased the expression levels of SOD and CAT, and decreased the expression level of MDA. The morphological examination showed that the ghrelin-treated lens organ culture maintained transparency. This is the first report to show that ghrelin can protect HLECs from H2O2-induced oxidative stress. Our findings suggest that ghrelin may prevent the progression of cataracts, which has treatment value for ophthalmologists.
Collapse
|
33
|
Ghrelin ameliorates acute lung injury induced by oleic acid via inhibition of endoplasmic reticulum stress. Life Sci 2017; 196:1-8. [PMID: 28751159 DOI: 10.1016/j.lfs.2017.07.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/14/2017] [Accepted: 07/21/2017] [Indexed: 12/31/2022]
Abstract
AIMS Acute lung injury (ALI) is associated with excessive mortality and lacks appropriate therapy. Ghrelin is a novel peptide that protects the lung against ALI. This study aimed to investigate whether endoplasmic reticulum stress (ERS) mediates the protective effect of ghrelin on ALI. MAIN METHODS We used a rat oleic acid (OA)-induced ALI model. Pulmonary impairment was detected by hematoxylin and eosin (HE) staining, lung mechanics, wet/dry weight ratio, and arterial blood gas analysis. Plasma and lung content of ghrelin was examined by ELISA, and mRNA expression was measured by quantitative real-time PCR. Protein levels were detected by western blot. KEY FINDINGS Rats with OA treatment showed significant pulmonary injury, edema, inflammatory cellular infiltration, cytokine release, hypoxia and CO2 retention as compared with controls. Plasma and pulmonary content of ghrelin was reduced in rats with ALI, and mRNA expression was downregulated. Ghrelin (10nmol/kg) treatment ameliorated the above symptoms, but treatment with the ghrelin antagonists D-Lys3 GHRP-6 (1μmol/kg) and JMV 2959 (6mg/kg) exacerbated the symptoms. ERS induced by OA was prevented by ghrelin and augmented by ghrelin antagonist treatment. The ERS inducer, tunicamycin (Tm) prevented the ameliorative effect of ghrelin on ALI. The decreased ratio of p-Akt and Akt induced by OA was improved by ghrelin treatment, and was further exacerbated by ghrelin antagonists. SIGNIFICANCE Ghrelin protects against ALI by inhibiting ERS. These results provide a new target for prevention and therapy of ALI.
Collapse
|
34
|
Roh HT, Cho SY, So WY. Obesity promotes oxidative stress and exacerbates blood-brain barrier disruption after high-intensity exercise. JOURNAL OF SPORT AND HEALTH SCIENCE 2017; 6:225-230. [PMID: 30356585 PMCID: PMC6188985 DOI: 10.1016/j.jshs.2016.06.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/25/2016] [Accepted: 04/06/2016] [Indexed: 05/16/2023]
Abstract
PURPOSE The purpose of this study was to investigate the effects of obesity and high-intensity acute exercise on oxidant-antioxidant status, neurotrophic factor expression, and blood-brain barrier (BBB) disruption. METHODS Twenty-four healthy, untrained men (12 non-obese (mean 14.9% body fat) and 12 obese subjects (mean 29.8% body fat)) performed 20 min of continuous submaximal aerobic exercise at 85% maximal oxygen consumption. Blood sampling was performed to examine the oxidant-antioxidant status (reactive oxygen species (ROS) and superoxide dismutase (SOD)), neurotrophic factors (brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF)), and BBB disruption (S100β and neuron-specific enolase) before and after acute exercise. RESULTS The obese group showed significantly higher pre-exercise serum ROS levels and significantly lower pre-exercise serum SOD levels than the non-obese group (p < 0.05). Serum ROS, SOD, BDNF, NGF, and S100β levels were significantly increased post-exercise compared with pre-exercise levels in both the non-obese and the obese groups (p < 0.05). The obese group showed significantly higher serum ROS, BDNF, NGF, and S100β levels post-exercise compared to the non-obese group (p < 0.05). CONCLUSION Our study suggests that episodic vigorous exercise can increase oxidative stress and blood neurotrophic factor levels and induce disruption of the BBB. Moreover, high levels of neurotrophic factor in the blood after exercise in the obese group may be due to BBB disruption, and it is assumed that oxidative stress was the main cause of this BBB disruption.
Collapse
Affiliation(s)
- Hee-Tae Roh
- Department of Physical Education, Dong-A University, Busan 604-714, Republic of Korea
| | - Su-Youn Cho
- School of Taekwondo, College of Creative Human Resources, Youngsan University, Yangsan-si 626-790, Republic of Korea
| | - Wi-Young So
- Sports and Health Care Major, College of Humanities and Arts, Korea National University of Transportation, Chungju-si 380-702, Republic of Korea
- Corresponding author.
| |
Collapse
|
35
|
Colldén G, Tschöp MH, Müller TD. Therapeutic Potential of Targeting the Ghrelin Pathway. Int J Mol Sci 2017; 18:ijms18040798. [PMID: 28398233 PMCID: PMC5412382 DOI: 10.3390/ijms18040798] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Ghrelin was discovered in 1999 as the endogenous ligand of the growth-hormone secretagogue receptor 1a (GHSR1a). Since then, ghrelin has been found to exert a plethora of physiological effects that go far beyond its initial characterization as a growth hormone (GH) secretagogue. Among the numerous well-established effects of ghrelin are the stimulation of appetite and lipid accumulation, the modulation of immunity and inflammation, the stimulation of gastric motility, the improvement of cardiac performance, the modulation of stress, anxiety, taste sensation and reward-seeking behavior, as well as the regulation of glucose metabolism and thermogenesis. Due to a variety of beneficial effects on systems’ metabolism, pharmacological targeting of the endogenous ghrelin system is widely considered a valuable approach to treat metabolic complications, such as chronic inflammation, gastroparesis or cancer-associated anorexia and cachexia. The aim of this review is to discuss and highlight the broad pharmacological potential of ghrelin pathway modulation for the treatment of anorexia, cachexia, sarcopenia, cardiopathy, neurodegenerative disorders, renal and pulmonary disease, gastrointestinal (GI) disorders, inflammatory disorders and metabolic syndrome.
Collapse
Affiliation(s)
- Gustav Colldén
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany.
| | - Timo D Müller
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
- Institute for Diabetes and Obesity (IDO), Business Campus Garching-Hochbrück, Parkring 13, 85748 Garching, Germany.
| |
Collapse
|
36
|
Koyuncuoğlu T, Vızdıklar C, Üren D, Yılmaz H, Yıldırım Ç, Atal SS, Akakın D, Kervancıoğlu Demirci E, Yüksel M, Yeğen BÇ. Obestatin improves oxidative brain damage and memory dysfunction in rats induced with an epileptic seizure. Peptides 2017; 90:37-47. [PMID: 28223092 DOI: 10.1016/j.peptides.2017.02.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/08/2017] [Accepted: 02/16/2017] [Indexed: 11/16/2022]
Abstract
Obestatin was shown to alleviate renal, gastrointestinal and haemorrhage-induced brain injury in rats. In order to investigate the neuroprotective effects of obestatin on seizure-induced oxidative brain injury, an epileptic seizure was induced with a single intraperitoneal (i.p.) dose of pentylenetetrazole (PTZ, 45mg/kg) in male Wistar rats. Thirty minutes before the PTZ injection, rats were treated with either saline or obestatin (1μg/kg, i.p.). Seizure was video-taped and then evaluated by using Racine's scoring (0-5). For the assessment of memory function, passive-avoidance test was performed before seizure induction, which was repeated on the 3rd day of seizure. The rats were decapitated at the 24th or 72nd hour of seizures and brain tissues were obtained for histopathological examination and for measuring levels of malondialdehyde (MDA), glutathione (GSH), reactive oxygen radicals and myeloperoxidase (MPO) activity. Obestatin treatment reduced the average seizure score, decreased the occurrence and duration of generalized tonic-clonic seizures, presenting with a shorter latency to their onset. Increased lipid peroxidation and enhanced generation of oxygen-derived radicals detected at the post-seizure 72nd h were suppressed by the consecutive treatments of obestatin, but no changes were observed by the single obestatin treatment in the 24-h seizure group. Neuronal damage and increased GFAP immunoreactivity, observed in the hippocampal areas and cortex of PTZ-induced rats were alleviated in 3-day obestatin-treated PTZ group. PTZ-induced memory dysfunction was significantly improved in obestatin-treated PTZ group as compared to saline-treated rats. The present data indicate that obestatin ameliorated the severity of PTZ-induced seizures, improved memory dysfunction and reduced neuronal damage by limiting oxidative damage.
Collapse
Affiliation(s)
| | - Caner Vızdıklar
- Marmara University School of Medicine, Department of Physiology, Turkey
| | - Doğan Üren
- Marmara University School of Medicine, Department of Physiology, Turkey
| | - Hakan Yılmaz
- Marmara University School of Medicine, Department of Physiology, Turkey
| | - Çağan Yıldırım
- Marmara University School of Medicine, Department of Physiology, Turkey
| | - Sefa Semih Atal
- Marmara University School of Medicine, Department of Physiology, Turkey
| | - Dilek Akakın
- Marmara University School of Medicine, Department of Histology and Embryology, Turkey
| | | | - Meral Yüksel
- Marmara University Vocational School of Health Related Professions, Istanbul, Turkey
| | - Berrak Ç Yeğen
- Marmara University School of Medicine, Department of Physiology, Turkey.
| |
Collapse
|
37
|
Xu J, Xu Z, Yan A. Prostaglandin E2 EP4 Receptor Activation Attenuates Neuroinflammation and Early Brain Injury Induced by Subarachnoid Hemorrhage in Rats. Neurochem Res 2017; 42:1267-1278. [PMID: 28239768 PMCID: PMC5375972 DOI: 10.1007/s11064-016-2168-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/19/2016] [Accepted: 12/26/2016] [Indexed: 01/08/2023]
Abstract
Activation of E prostanoid 4 receptor (EP4) shows neuroprotective effects in multiple central nervous system (CNS) lesions, but the roles of EP4 receptor in subarachnoid hemorrhage (SAH) are not explored. This study was designed to research the effects of EP4 modulation on early brain injury (EBI) after experimental SAH in rats. We found that the administration of EP4 selective agonist AE1-329 significantly improved neurological dysfunction, blood brain barrier (BBB) damage and brain edema at 24 h after SAH. Furthermore, AE1-329 obviously reduced the number of activated microglia and the mRNA and protein levels of pro-inflammatory cytokines, and increased Ser1177 phosphorylated endothelial nitric oxide synthase (Ser1177 p-eNOS). Moreover, AE1-329 significantly reduced the number of TUNEL-positive cells and active caspase-3 in cortex after SAH. The EP4 selective antagonist AE3-208 was also administrated and the opposite effects were achieved. Our results indicate that activation of EP4 protects brain from EBI through downregulating neuroinflammation reaction after SAH.
Collapse
Affiliation(s)
- Jie Xu
- Department of Neurosurgery, Huzhou Central Hospital, 198 Hongqi Lane, Huzhou, 313003, China
| | - Zhen Xu
- Department of Neurosurgery, First Affiliated Hospital of Zhejiang Chinese Medicine University, 54 Youdian Lane, Hangzhou, 310006, China
| | - Ai Yan
- Department of Neurosurgery, Huzhou Central Hospital, 198 Hongqi Lane, Huzhou, 313003, China.
| |
Collapse
|
38
|
Pereira JADS, da Silva FC, de Moraes-Vieira PMM. The Impact of Ghrelin in Metabolic Diseases: An Immune Perspective. J Diabetes Res 2017; 2017:4527980. [PMID: 29082258 PMCID: PMC5610818 DOI: 10.1155/2017/4527980] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/07/2017] [Accepted: 07/31/2017] [Indexed: 01/22/2023] Open
Abstract
Obesity and insulin resistance have reached epidemic proportions. Obesogenic conditions are associated with increased risk for the development of other comorbidities and obesity-related diseases. In metabolic disorders, there is chronic low-grade inflammation induced by the activation of immune cells, especially in metabolic relevant organs such as white adipose tissue (WAT). These immune cells are regulated by environmental and systemic cues. Ghrelin is a peptide secreted mainly by X/A-like gastric cells and acts through the growth hormone secretagogue receptor (GHS-R). This receptor is broadly expressed in the central nervous system (CNS) and in several cell types, including immune cells. Studies show that ghrelin induces an orexigenic state, and there is increasing evidence implicating an immunoregulatory role for ghrelin. Ghrelin mainly acts on the innate and adaptive immune systems to suppress inflammation and induce an anti-inflammatory profile. In this review, we discuss the immunoregulatory roles of ghrelin, the mechanisms by which ghrelin acts and potential pharmacological applications for ghrelin in the treatment of obesity-associated inflammatory diseases, such as type 2 diabetes (T2D).
Collapse
Affiliation(s)
- Jéssica Aparecida da Silva Pereira
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| | - Felipe Corrêa da Silva
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
| | - Pedro Manoel Mendes de Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
39
|
Jiao Q, Du X, Li Y, Gong B, Shi L, Tang T, Jiang H. The neurological effects of ghrelin in brain diseases: Beyond metabolic functions. Neurosci Biobehav Rev 2016; 73:98-111. [PMID: 27993602 DOI: 10.1016/j.neubiorev.2016.12.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 12/01/2016] [Accepted: 12/10/2016] [Indexed: 02/08/2023]
Abstract
Ghrelin, a peptide released by the stomach that plays a major role in regulating energy metabolism, has recently been shown to have effects on neurobiological behaviors. Ghrelin enhances neuronal survival by reducing apoptosis, alleviating inflammation and oxidative stress, and accordingly improving mitochondrial function. Ghrelin also stimulates the proliferation, differentiation and migration of neural stem/progenitor cells (NS/PCs). Additionally, the ghrelin is benefit for the recovery of memory, mood and cognitive dysfunction after stroke or traumatic brain injury. Because of its neuroprotective and neurogenic roles, ghrelin may be used as a therapeutic agent in the brain to combat neurodegenerative disease. In this review, we highlight the pre-clinical evidence and the proposed mechanisms underlying the role of ghrelin in physiological and pathological brain function.
Collapse
Affiliation(s)
- Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Yong Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Bing Gong
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China.
| | - Limin Shi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Tingting Tang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| |
Collapse
|
40
|
Liu Z, Yu T, Yang H, Tian X, Feng L. WITHDRAWN: Decreased level of endogenous ghrelin is involved in the progression of lung injury induced by oleic acid. Life Sci 2016:S0024-3205(16)30675-0. [PMID: 27894854 DOI: 10.1016/j.lfs.2016.11.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 11/15/2016] [Accepted: 11/24/2016] [Indexed: 01/18/2023]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Zhijun Liu
- Department of Respiration, Liaocheng People's Hospital, Liaocheng 252000, China.
| | - Ting Yu
- Department of Ultrasound, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Haitao Yang
- Department of Respiration, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Xiuli Tian
- Department of Respiration, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Linlin Feng
- Department of Respiration, Liaocheng People's Hospital, Liaocheng 252000, China
| |
Collapse
|
41
|
Zhong YW, Wu J, Hu HL, Li WX, Zhong Y. Protective effect 3,4-dihydroxyphenylethanol in subarachnoid hemorrhage provoked oxidative neuropathy. Exp Ther Med 2016; 12:1908-1914. [PMID: 27588109 DOI: 10.3892/etm.2016.3526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/22/2016] [Indexed: 12/25/2022] Open
Abstract
Clinical studies have indicated that early brain injury (EBI) following subarachnoid hemorrhage (SAH) is associated with fatal outcomes. Oxidative stress and brain edema are the characteristic pathological events in occurrence EBI following SAH. The present study aimed to examine the effect of 3,4-dihydroxyphenylethanol (DOPET) against SAH-induced EBI, and to demonstrate whether the effect is associated with its potent free radical scavenging property. SAH was induced in rats using an endovascular perforation technique, and 24 h later the rats displayed diminished neurological scores and brain edema. Furthermore, elevated malondialdehyde (an index of lipid peroxidation) and depleted levels of antioxidants were observed in the rat cerebral cortex tissue. Quantitative polymerase chain reaction analysis indicated the upregulated mRNA expression of the apoptotic markers caspase-3 and -9 in the cerebral cortex. Furthermore, the protein expression levels of the proinflammatory cytokines tumor necrosis factor-α, interleukin (IL)-1β and IL-6 were significantly upregulated in SAH-induced rats. By constrast, treatment with DOPET significantly attenuated EBI by reducing brain edema, elevation of antioxidant status, inhibition of apoptosis and inflammation. In this context, DOPET may be a potent agent in the treatment of EBI following SAH, as a result of its free radical scavenging capacity.
Collapse
Affiliation(s)
- Yu-Wen Zhong
- Department of Neurology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| | - Juan Wu
- Department of Obstetrics and Gynecology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| | - Hua-Long Hu
- Department of Neurology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| | - Wei-Xin Li
- Department of Neurology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| | - Yong Zhong
- Department of Neurology, Tongcheng People's Hospital, Xianning, Hubei 437400, P.R. China
| |
Collapse
|
42
|
Ghrelin attenuates brain injury in septic mice via PI3K/Akt signaling activation. Brain Res Bull 2016; 124:278-85. [DOI: 10.1016/j.brainresbull.2016.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/24/2016] [Accepted: 06/06/2016] [Indexed: 01/14/2023]
|
43
|
Abstract
Poor neurological outcome remains a major problem in patients with cardiac arrest. Ghrelin has been shown to be neuroprotective in models of neurologic injury in vitro and in vivo. This study was performed to assess the effects of ghrelin on postresuscitation brain injury in a rat model of cardiac arrest. Sprague-Dawley rats were subjected to 6-min cardiac arrest and resuscitated successfully. Either vehicle (saline) or ghrelin (80 μg/kg) was injected blindly immediately after return of spontaneous circulation (ROSC). A tape removal test was performed to evaluate neurological function at 24, 48, and 72 h after ROSC. Then, brain tissues were harvested and coronal brain sections were analyzed by hematoxylin and eosin (HE) staining for neuronal viability and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining for apoptosis in hippocampal CA1 sectors. In additional groups, rats were sacrificed at 6 h after ROSC, and hippocampal tissues were collected for further analysis. We found that animals treated with ghrelin had improved neurological performances, reduced neuronal injury, and inhibited neuronal apoptosis compared with the vehicle group. Moreover, ghrelin treatment was associated with the following: (1) a decrease in caspase-3 up-regulation and an increased Bcl-2/Bax ratio, (2) a reduction in maleic dialdehyde content and an up-regulation in superoxide dismutase activity, and (3) an increase in uncoupling protein 2 (UCP-2) expression. Our results suggest that ghrelin treatment attenuated postresuscitation brain injury in rats, possibly via regulation of apoptosis, oxidative stress, and mitochondrial UCP-2 expression. Ghrelin may have therapeutic potential when administered after cardiac arrest and cardiopulmonary resuscitation.
Collapse
|
44
|
The functional and structural changes in the basilar artery due to overpressure blast injury. J Cereb Blood Flow Metab 2015; 35:1950-6. [PMID: 26104291 PMCID: PMC4671114 DOI: 10.1038/jcbfm.2015.151] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/13/2015] [Accepted: 05/07/2015] [Indexed: 12/13/2022]
Abstract
Overpressure blast-wave induced brain injury (OBI) leads to progressive pathophysiologic changes resulting in a reduction in brain blood flow, blood brain barrier breakdown, edema, and cerebral ischemia. The aim of this study was to evaluate cerebral vascular function after single and repeated OBI. Male Sprague-Dawley rats were divided into three groups: Control (Naive), single OBI (30 psi peak pressure, 1 to 2 msec duration), and repeated (days 1, 4, and 7) OBI (r-OBI). Rats were killed 24 hours after injury and the basilar artery was isolated, cannulated, and pressurized (90 cm H2O). Vascular responses to potassium chloride (KCl) (30 to 100 mmol/L), endothelin-1 (10(-12) to 10(-7) mol/L), acetylcholine (ACh) (10(-10) to 10(-4) mol/L) and diethylamine-NONO-ate (DEA-NONO-ate) (10(-10) to 10(-4) mol/L) were evaluated. The OBI resulted in an increase in the contractile responses to endothelin and a decrease in the relaxant responses to ACh in both single and r-OBI groups. However, impaired DEA-NONO-ate-induced vasodilation and increased wall thickness to lumen ratio were observed only in the r-OBI group. The endothelin-1 type A (ET(A)) receptor and endothelial nitric oxide synthase (eNOS) immunoreactivity were significantly enhanced by OBI. These findings indicate that both single and r-OBI impairs cerebral vascular endothelium-dependent dilation, potentially a consequence of endothelial dysfunction and/or vascular remodelling in basilar arteries after OBI.
Collapse
|
45
|
Price L, Wilson C, Grant G. Blood–Brain Barrier Pathophysiology following Traumatic Brain Injury. TRANSLATIONAL RESEARCH IN TRAUMATIC BRAIN INJURY 2015. [DOI: 10.1201/b18959-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Shi SS, Zhang HB, Wang CH, Yang WZ, Liang RS, Chen Y, Tu XK. Propofol Attenuates Early Brain Injury After Subarachnoid Hemorrhage in Rats. J Mol Neurosci 2015; 57:538-45. [PMID: 26342279 DOI: 10.1007/s12031-015-0634-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 07/28/2015] [Indexed: 02/01/2023]
Abstract
Our previous studies demonstrated that propofol protects rat brain against focal cerebral ischemia. However, whether propofol attenuates early brain injury after subarachnoid hemorrhage in rats remains unknown until now. The present study was performed to evaluate the effect of propofol on early brain injury after subarachnoid hemorrhage in rats and further explore the potential mechanisms. Sprague-Dawley rats underwent subarachnoid hemorrhage (SAH) by endovascular perforation then received treatment with propofol (10 or 50 mg/kg) or vehicle after 2 and 12 h of SAH. SAH grading, neurological scores, brain water content, Evans blue extravasation, the myeloperoxidase activity, and malondialdehyde (MDA) content were measured 24 h after SAH. Expression of nuclear factor erythroid-related factor 2 (Nrf2), nuclear factor-kappa B (NF-κB) p65, and aquaporin 4 (AQP4) expression in rat brain were detected by Western blot. Expression of cyclooxygenase-2 (COX-2) and matrix metalloproteinase-9 (MMP-9) were determined by reverse transcription-polymerase chain reaction (RT-PCR). Expressions of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were assessed by ELISA. Neurological scores, brain water content, Evans blue extravasation, the myeloperoxidase activity, and MDA content were significantly reduced by propofol. Furthermore, expression of Nrf2 in rat brain was upregulated by propofol, and expression of NF-κB p65, AQP4, COX-2, MMP-9, TNF-α, and IL-1β in rat brain were attenuated by propofol. Our results demonstrated that propofol improves neurological scores, reduces brain edema, blood-brain barrier (BBB) permeability, inflammatory reaction, and lipid peroxidation in rats of SAH. Propofol exerts neuroprotection against SAH-induced early brain injury, which might be associated with the inhibition of inflammation and lipid peroxidation.
Collapse
Affiliation(s)
- Song-sheng Shi
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Hua-bin Zhang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Chun-hua Wang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Wei-zhong Yang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Ri-sheng Liang
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Ye Chen
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Xian-kun Tu
- Department of Neurosurgery, The Affiliated Union Hospital of Fujian Medical University, Neurosurgery Research Institute of Fujian Province, No. 29 Xinquan Road, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
47
|
Şen LS, Karakoyun B, Yeğen C, Akkiprik M, Yüksel M, Ercan F, Özer A, Yeğen BÇ. Treatment with either obestatin or ghrelin attenuates mesenteric ischemia-reperfusion-induced oxidative injury of the ileum and the remote organ lung. Peptides 2015; 71:8-19. [PMID: 26032330 DOI: 10.1016/j.peptides.2015.04.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/06/2015] [Accepted: 04/14/2015] [Indexed: 12/14/2022]
Abstract
To evaluate the effects of exogenous ghrelin or obestatin on intestinal injury and accompanying pulmonary injury, intestinal ischemia-reperfusion (I/R) was induced in rats by obstructing the superior mesenteric artery for 60min, whereas laparotomy was performed in the sham group. At the beginning of the 90-min reperfusion period, the rats were injected with obestatin (100μg/kg), ghrelin (10ng/kg), or saline intravenously (iv). At the end of reperfusion, the blood, ileum, and lung samples were taken for the histological and biochemical assays. In the saline-treated I/R group, the increased serum interleukin (IL)-1β level, high damage scores, and elevated tissue malondialdehyde level and collagen content in both tissues were significantly reduced by obestatin or ghrelin. Increased ileal myeloperoxidase activity of the saline-treated I/R group was reduced by treatment with obestatin or ghrelin, whereas increased pulmonary myeloperoxidase activity was reduced with administration of obestatin. Increased DNA fragmentation in the ileum of the saline-treated I/R group was reduced by both peptides. Elevated luminol-lucigenin chemiluminescence levels and nuclear factor kappa B (NF-κB) messenger RNA (mRNA) expression in the ileum of the saline-treated-I/R group were significantly decreased by obestatin or ghrelin treatment. I/R-induced depletion of the antioxidant glutathione in both ileal and pulmonary tissues was prevented with either obestatin or ghrelin treatment. Administration of either obestatin or ghrelin exerts similar protective effects against I/R-induced ileal and pulmonary injury, thus warranting further investigation for their possible use against ischemic intestinal injury.
Collapse
Affiliation(s)
- Leyla Semiha Şen
- Marmara University School of Medicine, Department of General Surgery, Istanbul, Turkey
| | - Berna Karakoyun
- Marmara University Faculty of Health Sciences, Department of Basic Health Sciences, Istanbul, Turkey
| | - Cumhur Yeğen
- Marmara University School of Medicine, Department of General Surgery, Istanbul, Turkey
| | - Mustafa Akkiprik
- Marmara University School of Medicine, Department of Medical Biology, Istanbul, Turkey
| | - Meral Yüksel
- Marmara University Vocational School of Health Related Professions, Department of Medical Laboratory, Istanbul, Turkey
| | - Feriha Ercan
- Marmara University School of Medicine, Department of Histology and Embryology, Istanbul, Turkey
| | - Ayşe Özer
- Marmara University School of Medicine, Department of Medical Biology, Istanbul, Turkey
| | - Berrak Ç Yeğen
- Marmara University School of Medicine, Department of Physiology, Istanbul, Turkey.
| |
Collapse
|
48
|
Tian Y, Guo SX, Li JR, Du HG, Wang CH, Zhang JM, Wu Q. Topiramate attenuates early brain injury following subarachnoid haemorrhage in rats via duplex protection against inflammation and neuronal cell death. Brain Res 2015; 1622:174-85. [PMID: 26086367 DOI: 10.1016/j.brainres.2015.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 06/04/2015] [Accepted: 06/07/2015] [Indexed: 01/31/2023]
Abstract
Early brain injury (EBI) following aneurysmal subarachnoid haemorrhage (SAH) insults contributes to the poor prognosis and high mortality observed in SAH patients. Topiramate (TPM) is a novel, broad-spectrum, antiepileptic drug with a reported protective effect against several brain injuries. The current study aimed to investigate the potential of TPM for neuroprotection against EBI after SAH and the possible dose-dependency of this effect. An endovascular perforation SAH model was established in rats, and TPM was administered by intraperitoneal injection after surgery at three different doses (20mg/kg, 40mg/kg, and 80mg/kg). The animals' neurological scores and brain water content were evaluated, and ELISA, Western blotting and immunostaining assays were conducted to assess the effect of TPM. The results revealed that TPM lowers the elevated levels of myeloperoxidase and proinflammatory mediators observed after SAH in a dose-related fashion, and the nuclear factor-kappa B (NF-κB) signalling pathway is the target of neuroinflammation regulation. In addition, TPM ameliorated SAH-induced cortical neuronal apoptosis by influencing Bax, Bcl-2 and cleaved caspase-3 protein expression, and the effect of TPM was enhanced in a dose-dependent manner. Various dosages of TPM also upregulated the protein expression of the γ-aminobutyric acid (GABA)-ergic signalling molecules, GABAA receptor (GABAAR) α1, GABAAR γ2, and K(+)-Cl(-) co-transporter 2 (KCC2) together and downregulated Na(+)-K(+)-Cl(-) co-transporter 1 (NKCC1) expression. Thus, TPM may be an effective neuroprotectant in EBI after SAH by regulating neuroinflammation and neuronal cell death.
Collapse
Affiliation(s)
- Yong Tian
- Department of Neurosurgery, Second Affiliated Hospital, Zhejiang Chinese Medical University, 318 Chaowang Road, Hangzhou 310005, Zhejiang, China; Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Song-Xue Guo
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China; Department of Burns, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Jian-Ru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Hang-Gen Du
- Department of Neurosurgery, Second Affiliated Hospital, Zhejiang Chinese Medical University, 318 Chaowang Road, Hangzhou 310005, Zhejiang, China
| | - Chao-Hui Wang
- Department of Neurosurgery, Ruian People's Hospital, 108 Wansong Road, Ruian 325200, Zhejiang, China
| | - Jian-Min Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China
| | - Qun Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang, China.
| |
Collapse
|
49
|
Can N, Catak O, Turgut B, Demir T, Ilhan N, Kuloglu T, Ozercan IH. Neuroprotective and antioxidant effects of ghrelin in an experimental glaucoma model. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2819-29. [PMID: 26082612 PMCID: PMC4459614 DOI: 10.2147/dddt.s83067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Damage to retinal ganglion cells due to elevation of intraocular pressure (IOP) is responsible for vision loss in glaucoma. Given that loss of these cells is irreversible, neuroprotection is crucial in the treatment of glaucoma. In this study, we investigated the possible antioxidant and neuroprotective effects of ghrelin on the retina in an experimental glaucoma model. Twenty-one Sprague–Dawley rats were randomly assigned to three groups comprising seven rats each. The rats in the control group were not operated on and did not receive any treatment. In all rats in the other groups, IOP was increased by cauterization of the limbal veins. After creation of the IOP increase, saline 1 mL/kg or ghrelin 40 μg/kg was administered intraperitoneally every day for 14 days in the vehicle control group and ghrelin groups, respectively. On day 14 of the study, the eyes were enucleated. Levels of malondialdehyde (MDA), nitric oxide (NO), and nitric oxide synthase-2 (NOS2) in anterior chamber fluid were measured. The retinas were subjected to immunohistochemistry staining for glial fibrillary acidic protein (GFAP), S-100, and vimentin expression. Mean levels of MDA, NO, and NOS2 in the aqueous humor were higher in the vehicle control group than in the control group (P<0.05). Mean levels of MDA, NO, and NOS2 in the ghrelin group did not show a significant increase compared with levels in the control group (P>0.05). Retinal TUNEL and immunohistochemistry staining in the vehicle control group showed an increase in apoptosis and expression of GFAP, S-100, and vimentin compared with the control group (P<0.05). In the ghrelin group, apoptosis and expression of GFAP, S-100, and vimentin was significantly lower than in the vehicle control group (P<0.05). This study suggests that ghrelin has antioxidant and neuroprotective effects on the retina in an experimental glaucoma model.
Collapse
Affiliation(s)
- Nagehan Can
- Department of Ophthalmology, Elazığ Training and Research Hospital, Fırat University, Elazığ, Turkey
| | - Onur Catak
- Department of Ophthalmology, School of Medicine, Fırat University, Elazığ, Turkey
| | - Burak Turgut
- Department of Ophthalmology, School of Medicine, Fırat University, Elazığ, Turkey
| | - Tamer Demir
- Department of Ophthalmology, School of Medicine, Fırat University, Elazığ, Turkey
| | - Nevin Ilhan
- Department of Biochemistry, School of Medicine, Fırat University, Elazığ, Turkey
| | - Tuncay Kuloglu
- Department of Histology and Embryology, School of Medicine, Fırat University, Elazığ, Turkey
| | | |
Collapse
|
50
|
Aygen B, Kucuksu M, Aydin S, Ozercan IH. Effect of enalapril maleate on ghrelin levels in metabolic syndrome in rats. Peptides 2015; 67:39-44. [PMID: 25784288 DOI: 10.1016/j.peptides.2015.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/05/2015] [Accepted: 03/05/2015] [Indexed: 12/13/2022]
Abstract
We have explored how enalapril affects ghrelin levels in serum and renal tissues of rats with fructose-induced MetS, using 5-week-old Wistar albino male rats weighing 220 ± 20 g. They divided into 5 groups: (i) control (CT), no fructose supplement fed on standard rat pellet and tap water for 60 days, (ii) metabolic syndrome (MetS) fed with 10% fructose for 60 days, (iii) rats after metabolic syndrome developed treated with enalapril over 30 days (MetS+E30), (iv) rats in which only enalapril was administered for 60 days (E60), and (v) MetS-treated with enalapril for 60 days (MetS+E60). Enalapril maleate was given at 20mg/kg per day by gavage. Fasting serum insulin, uric acid, triglyceride, low-density lipoprotein cholesterol and total cholesterol levels were significantly higher, and the amount of high density lipoprotein cholesterol, and acylated and desacyl ghrelin levels was significantly lower in the MetS groups. Ghrelins were significantly lower in all 3 groups, which were administered enalapril than that of MetS and the control group. Immunohistochemical staining showed that the density of ghrelin was parallel to the serum levels of the peptide. Ghrelin immunoreactivity in the kidneys was of moderate density in the distal and collecting tubules, mild density in the proximal tubule and glomeruli, whereas the density decreased in the MetS group and other enalapril-treated groups. In conclusion, ghrelin levels in MetS groups were significantly lower than control group, and thus Enalapril treatment improves components of MetS and has direct effects on serum ghrelin levels that are independent of MetS.
Collapse
Affiliation(s)
- Bilge Aygen
- Department of Nephrology, Firat University, Faculty of Medicine, Elazig, 23119, Turkey.
| | - Mehmet Kucuksu
- Department of Nephrology, Firat University, Faculty of Medicine, Elazig, 23119, Turkey
| | - Suleyman Aydin
- Department of Medical Biochemistry (Firat Hormones Research Group), Firat University, Faculty of Medicine, Elazig, 23119, Turkey.
| | - Ibrahim Hanifi Ozercan
- Department of Medical Pathology, Firat University, Faculty of Medicine, Elazig, 23119, Turkey
| |
Collapse
|