1
|
Meng K, Liu Q, Qin Y, Qin W, Zhu Z, Sun L, Jiang M, Adu-Amankwaah J, Gao F, Tan R, Yuan J. Mechanism of mitochondrial oxidative phosphorylation disorder in male infertility. Chin Med J (Engl) 2025; 138:379-388. [PMID: 38855875 PMCID: PMC11845199 DOI: 10.1097/cm9.0000000000003126] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Indexed: 06/11/2024] Open
Abstract
ABSTRACT Male infertility has become a global concern, accounting for 20-70% of infertility. Dysfunctional spermatogenesis is the most common cause of male infertility; thus, treating abnormal spermatogenesis may improve male infertility and has attracted the attention of the medical community. Mitochondria are essential organelles that maintain cell homeostasis and normal physiological functions in various ways, such as mitochondrial oxidative phosphorylation (OXPHOS). Mitochondrial OXPHOS transmits electrons through the respiratory chain, synthesizes adenosine triphosphate (ATP), and produces reactive oxygen species (ROS). These mechanisms are vital for spermatogenesis, especially to maintain the normal function of testicular Sertoli cells and germ cells. The disruption of mitochondrial OXPHOS caused by external factors can result in inadequate cellular energy supply, oxidative stress, apoptosis, or ferroptosis, all inhibiting spermatogenesis and damaging the male reproductive system, leading to male infertility. This article summarizes the latest pathological mechanism of mitochondrial OXPHOS disorder in testicular Sertoli cells and germ cells, which disrupts spermatogenesis and results in male infertility. In addition, we also briefly outline the current treatment of spermatogenic malfunction caused by mitochondrial OXPHOS disorders. However, relevant treatments have not been fully elucidated. Therefore, targeting mitochondrial OXPHOS disorders in Sertoli cells and germ cells is a research direction worthy of attention. We believe this review will provide new and more accurate ideas for treating male infertility.
Collapse
Affiliation(s)
- Kai Meng
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, Shandong 272067, China
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong 272067, China
| | - Qian Liu
- College of Basic Medical, Jining Medical University, Jining, Shandong 272067, China
| | - Yiding Qin
- College of Basic Medical, Jining Medical University, Jining, Shandong 272067, China
| | - Wenjie Qin
- College of Second Clinical Medicine, Jining Medical University, Jining, Shandong 272067, China
| | - Ziming Zhu
- College of Second Clinical Medicine, Jining Medical University, Jining, Shandong 272067, China
| | - Longlong Sun
- College of Second Clinical Medicine, Jining Medical University, Jining, Shandong 272067, China
| | - Mingchao Jiang
- College of Second Clinical Medicine, Jining Medical University, Jining, Shandong 272067, China
| | - Joseph Adu-Amankwaah
- College of Basic Medical, Xuzhou Medical University, Xuzhou, Zhejiang 221004, China
| | - Fei Gao
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, Shandong 272067, China
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong 272067, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 101408, China
| | - Rubin Tan
- College of Basic Medical, Xuzhou Medical University, Xuzhou, Zhejiang 221004, China
| | - Jinxiang Yuan
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, Shandong 272067, China
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong 272067, China
| |
Collapse
|
2
|
Daimiel Naranjo I, Bhowmik A, Basukala D, Lo Gullo R, Mazaheri Y, Kapetas P, Eskreis-Winkler S, Pinker K, Thakur SB. Assessment of Hypoxia in Breast Cancer: Emerging Functional MR Imaging and Spectroscopy Techniques and Clinical Applications. J Magn Reson Imaging 2025; 61:83-96. [PMID: 38703143 DOI: 10.1002/jmri.29424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
Breast cancer is one of the most prevalent forms of cancer affecting women worldwide. Hypoxia, a condition characterized by insufficient oxygen supply in tumor tissues, is closely associated with tumor aggressiveness, resistance to therapy, and poor clinical outcomes. Accurate assessment of tumor hypoxia can guide treatment decisions, predict therapy response, and contribute to the development of targeted therapeutic interventions. Over the years, functional magnetic resonance imaging (fMRI) and magnetic resonance spectroscopy (MRS) techniques have emerged as promising noninvasive imaging options for evaluating hypoxia in cancer. Such techniques include blood oxygen level-dependent (BOLD) MRI, oxygen-enhanced MRI (OE) MRI, chemical exchange saturation transfer (CEST) MRI, and proton MRS (1H-MRS). These may help overcome the limitations of the routinely used dynamic contrast-enhanced (DCE) MRI and diffusion-weighted imaging (DWI) techniques, contributing to better diagnosis and understanding of the biological features of breast cancer. This review aims to provide a comprehensive overview of the emerging functional MRI and MRS techniques for assessing hypoxia in breast cancer, along with their evolving clinical applications. The integration of these techniques in clinical practice holds promising implications for breast cancer management. EVIDENCE LEVEL: 5 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Isaac Daimiel Naranjo
- Department of Radiology, HM Hospitales, Madrid, Spain
- School of Medicine, Universidad CEU San Pablo, Madrid, Spain
| | - Arka Bhowmik
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dibash Basukala
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), Center for Biomedical Imaging, NYU Langone Health, New York, New York, USA
| | - Roberto Lo Gullo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Yousef Mazaheri
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Panagiotis Kapetas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Sarah Eskreis-Winkler
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Katja Pinker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sunitha B Thakur
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
3
|
Akanchise T, Angelova A. Ginkgo Biloba and Long COVID: In Vivo and In Vitro Models for the Evaluation of Nanotherapeutic Efficacy. Pharmaceutics 2023; 15:pharmaceutics15051562. [PMID: 37242804 DOI: 10.3390/pharmaceutics15051562] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Coronavirus infections are neuroinvasive and can provoke injury to the central nervous system (CNS) and long-term illness consequences. They may be associated with inflammatory processes due to cellular oxidative stress and an imbalanced antioxidant system. The ability of phytochemicals with antioxidant and anti-inflammatory activities, such as Ginkgo biloba, to alleviate neurological complications and brain tissue damage has attracted strong ongoing interest in the neurotherapeutic management of long COVID. Ginkgo biloba leaf extract (EGb) contains several bioactive ingredients, e.g., bilobalide, quercetin, ginkgolides A-C, kaempferol, isorhamnetin, and luteolin. They have various pharmacological and medicinal effects, including memory and cognitive improvement. Ginkgo biloba, through its anti-apoptotic, antioxidant, and anti-inflammatory activities, impacts cognitive function and other illness conditions like those in long COVID. While preclinical research on the antioxidant therapies for neuroprotection has shown promising results, clinical translation remains slow due to several challenges (e.g., low drug bioavailability, limited half-life, instability, restricted delivery to target tissues, and poor antioxidant capacity). This review emphasizes the advantages of nanotherapies using nanoparticle drug delivery approaches to overcome these challenges. Various experimental techniques shed light on the molecular mechanisms underlying the oxidative stress response in the nervous system and help comprehend the pathophysiology of the neurological sequelae of SARS-CoV-2 infection. To develop novel therapeutic agents and drug delivery systems, several methods for mimicking oxidative stress conditions have been used (e.g., lipid peroxidation products, mitochondrial respiratory chain inhibitors, and models of ischemic brain damage). We hypothesize the beneficial effects of EGb in the neurotherapeutic management of long-term COVID-19 symptoms, evaluated using either in vitro cellular or in vivo animal models of oxidative stress.
Collapse
Affiliation(s)
- Thelma Akanchise
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| |
Collapse
|
4
|
Ma X, Xu J, Gao N, Tian J, Song T. Dexmedetomidine attenuates myocardial ischemia-reperfusion injury via inhibiting ferroptosis by the cAMP/PKA/CREB pathway. Mol Cell Probes 2023; 68:101899. [PMID: 36775106 DOI: 10.1016/j.mcp.2023.101899] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/14/2023]
Abstract
This study is to investigate the effects of dexmedetomidine on myocardial ischemia-reperfusion (I/R) injury and its molecular mechanisms. H9c2 cell injury model was constructed by the hypoxia/normoxia (H/R) conditions. Besides, cAMP response element-binding protein (CREB) overexpression and knockdown cell lines were constructed. Cell viability was determined by cell-counting kit 8. Biochemical assays were used to detect oxidative stress-related biomarkers, cell apoptosis, and ferroptosis-related markers. Our results showed that dexmedetomidine's protective effects on H/R-induced cell damage were reversed by the inhibition of protein kinase A (PKA), CREB, and extracellular signal regulated kinase 1/2 (ERK1/2). Treatment of dexmedetomidine ameliorated oxidative stress in the cardiomyocytes induced by H/R, whereas inhibition of PKA, CREB, or ERK1/2 reversed these protective effects. Cell death including cell necrosis, apoptosis, and ferroptosis was found in the cells under H/R insult. Interestingly, targeting CREB ameliorated ferroptosis and oxidative stress in these cells. In conclusion, dexmedetomidine attenuates myocardial I/R injury by suppressing ferroptosis through the cAMP/PKA/CREB signaling pathway.
Collapse
Affiliation(s)
- Xiaojing Ma
- Department of Anesthesiology, Shijiazhuang People's Hospital, Shijiazhuang, 050000, Hebei, China.
| | - Jia Xu
- Department of Anesthesiology, Shijiazhuang People's Hospital, Shijiazhuang, 050000, Hebei, China
| | - Nan Gao
- Department of Anesthesiology, Shijiazhuang People's Hospital, Shijiazhuang, 050000, Hebei, China
| | - Jun Tian
- Second Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, 050000, Hebei, China
| | - Tieying Song
- Department of Anesthesiology, Shijiazhuang People's Hospital, Shijiazhuang, 050000, Hebei, China
| |
Collapse
|
5
|
The secretory phenotypes of envenomed cells: Insights into venom cytotoxicity. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:193-230. [PMID: 36707202 DOI: 10.1016/bs.apcsb.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Snake envenomation is listed as Category A Neglected Tropical Diseases (NTD) by World Health Organization, indicates a severe public health problem. The global figures for envenomation cases are estimated to be more than 1.8 million annually. Even if the affected victims survive the envenomation, they might suffer from permanent morbidity due to local envenomation. One of the most prominent local envenomation is dermonecrosis. Dermonecrosis is a pathophysiological outcome of envenomation that often causes disability in the victims due to surgical amputations, deformities, contracture, and chronic ulceration. The key venom toxins associated with this local symptom are mainly attributed to substantial levels of enzymatic and non-enzymatic toxins as well as their possible synergistic actions. Despite so, the severity of the local tissue damage is based on macroscopic observation of the bite areas. Furthermore, limited knowledge is known about the key biomarkers involved in the pathogenesis of dermonecrosis. The current immunotherapy with antivenom is also ineffective against dermonecrosis. These local effects eventually end up as sequelae. There is also a global shortage of toxins-targeted therapeutics attributed to inadequate knowledge of the actual molecular mechanisms of cytotoxicity. This chapter discusses the characterization of secretory phenotypes of dermonecrosis as an advanced tool to indicate its severity and pathogenesis in envenomation. Altogether, the secretory phenotypes of envenomed cells and tissues represent the precise characteristics of dermonecrosis caused by venom toxins.
Collapse
|
6
|
Mitochondrial transplant after ischemia reperfusion promotes cellular salvage and improves lung function during ex-vivo lung perfusion. J Heart Lung Transplant 2023; 42:575-584. [PMID: 36707296 DOI: 10.1016/j.healun.2023.01.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 12/02/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND In lung transplantation, ischemia-reperfusion injury associated with mitochondrial damage can lead to graft rejection. Intact, exogenous mitochondria provide a unique treatment option to salvage damaged cells within lung tissue. METHODS We developed a novel method to freeze and store allogeneic mitochondria isolated from porcine heart tissue. Stored mitochondria were injected into a model of induced ischemia-reperfusion injury using porcine ex-vivo lung perfusion. Treatment benefits to immune modulation, antioxidant defense, and cellular salvage were evaluated. These findings were corroborated in human lungs undergoing ex-vivo lung perfusion. Lung tissue homogenate and primary lung endothelial cells were then used to address underlying mechanisms. RESULTS Following cold ischemia, mitochondrial transplant reduced lung pulmonary vascular resistance and tissue pro-inflammatory signaling and cytokine secretion. Further, exogenous mitochondria reduced reactive oxygen species by-products and promoted glutathione synthesis, thereby salvaging cell viability. These results were confirmed in a human model of ex-vivo lung perfusion wherein transplanted mitochondria decreased tissue oxidative and inflammatory signaling, improving lung function. We demonstrate that transplanted mitochondria induce autophagy and suggest that bolstered autophagy may act upstream of the anti-inflammatory and antioxidant benefits. Importantly, chemical inhibitors of the MEK autophagy pathway blunted the favorable effects of mitochondrial transplant. CONCLUSIONS These data provide direct evidence that mitochondrial transplant improves cellular health and lung function when administered during ex-vivo lung perfusion and suggest the mechanism of action may be through promotion of cellular autophagy. Data herein contribute new insights into the therapeutic potential of mitochondrial transplant to abate ischemia-reperfusion injury during lung transplant, and thus reduce graft rejection.
Collapse
|
7
|
Glutamine-dependent effects of nitric oxide on cancer cells subjected to hypoxia-reoxygenation. Nitric Oxide 2023; 130:22-35. [PMID: 36414197 DOI: 10.1016/j.niox.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/21/2022]
Abstract
Limited O2 availability can decrease essential processes in energy metabolism. However, cancers have developed distinct metabolic adaptations to these conditions. For example, glutaminolysis can maintain energy metabolism and hypoxia signaling. Additionally, it has been observed that nitric oxide (NO) possesses concentration-dependent, biphasic effects in cancer. NO has potent anti-tumor effects through modulating events such as angiogenesis and metastasis at low physiological concentrations and inducing cell death at higher concentrations. In this study, Ewing Sarcoma cells (A-673), MIA PaCa, and SKBR3 cells were treated with DetaNONOate (DetaNO) in a model of hypoxia (1% O2) and reoxygenation (21% O2). All 3 cell types showed NO-dependent inhibition of cellular O2 consumption which was enhanced as O2-tension decreased. L-Gln depletion suppressed the mitochondrial response to decreasing O2 tension in all 3 cell types and resulted in inhibition of Complex I activity. In A-673 cells the O2 tension dependent change in mitochondrial O2 consumption and increase in glycolysis was dependent on the presence of L-Gln. The response to hypoxia and Complex I activity were restored by α-ketoglutarate. NO exposure resulted in the A-673 cells showing greater sensitivity to decreasing O2 tension. Under conditions of L-Gln depletion, NO restored HIF-1α levels and the mitochondrial response to O2 tension possibly through the increase of 2-hydroxyglutarate. NO also resulted in suppression of cellular bioenergetics and further inhibition of Complex I which was not rescued by α-ketoglutarate. Taken together these data suggest that NO modulates the mitochondrial response to O2 differentially in the absence and presence of L-Gln. These data suggest a combination of metabolic strategies targeting glutaminolysis and Complex I in cancer cells.
Collapse
|
8
|
Galano M, Papadopoulos V. Role of Constitutive STAR in Mitochondrial Structure and Function in MA-10 Leydig Cells. Endocrinology 2022; 163:6608928. [PMID: 35704520 DOI: 10.1210/endocr/bqac091] [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: 03/26/2022] [Indexed: 11/19/2022]
Abstract
The steroidogenic acute regulatory protein (STAR; STARD1) is critical for the transport of cholesterol into the mitochondria for hormone-induced steroidogenesis. Steroidogenic cells express STAR under control conditions (constitutive STAR). On hormonal stimulation, STAR localizes to the outer mitochondrial membrane (OMM) where it facilitates cholesterol transport and where it is processed to its mature form. Here, we show that knockout of Star in MA-10 mouse tumor Leydig cells (STARKO1) causes defects in mitochondrial structure and function under basal conditions. We also show that overexpression of Star in STARKO1 cells exacerbates, rather than recovers, mitochondrial structure and function, which further disrupts the processing of STAR at the OMM. Our findings suggest that constitutive STAR is necessary for proper mitochondrial structure and function and that mitochondrial dysfunction leads to defective STAR processing at the OMM.
Collapse
Affiliation(s)
- Melanie Galano
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089, USA
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90089, USA
| |
Collapse
|
9
|
Siwicka-Gieroba D, Terpilowska S, Robba C, Barud M, Kubik-Komar A, Dabrowski W. The Connection Between Selected Caspases Levels in Bronchoalveolar Lavage Fluid and Severity After Brain Injury. Front Neurol 2022; 13:796238. [PMID: 35665033 PMCID: PMC9161272 DOI: 10.3389/fneur.2022.796238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Objective The interaction between the brain and lungs has been the subject of many clinical reports, while the exact impact of brain injury on the physiology of the respiratory system is still subject to numerous experimental studies. The purpose of this study was to investigate the activation of selected caspases levels in bronchoalveolar lavage fluid (mini BALF) of patients after isolated brain injury and their correlation with the severity of the injury. Methods The analysis was performed on patients who were admitted to the intensive care unit (ICU) for severe isolated brain injury from March 2018 to April 2020. All patients were intubated and mechanically ventilated. Mini BALF was collected within the first 6–8 h after trauma and on days 3 and 7 after admission. The concentrations of selected caspases were determined and correlated with the severity of brain injury evaluated by the Rotterdam CT Score, Glasgow Coma Score, and 28-day mortality. Results Our results showed significantly elevated levels of selected caspases on days 3 and 7 after brain injury, and revealed apoptosis activation during the first 7 days after brain trauma. We found a significant different correlation between the elevation of selected caspases 3, 6, 8, and 9, and the Glasgow Coma Score, Rotterdam CT scale, and 28-day mortality. Conclusions The increased levels of selected caspases in the mini BALF in our patients indicate an intensified activation of apoptosis in the lungs, which is related to brain injury itself via various apoptotic pathways and correlates with the severity of brain injury.
Collapse
Affiliation(s)
- Dorota Siwicka-Gieroba
- Department of Anaesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
- *Correspondence: Dorota Siwicka-Gieroba
| | | | - Chiara Robba
- Anaesthesia and Intensive Care, Policlinico San Martino, Deputy of the Neurointensive Care Section of European Society of Intensive Care Medicine, Genova, Italy
| | - Małgorzata Barud
- Department of Anaesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| | - Agnieszka Kubik-Komar
- Department of Applied Mathematics and Computer Science, University of Life Sciences in Lublin, Lublin, Poland
| | - Wojciech Dabrowski
- Department of Anaesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
10
|
Xin Y, Yang Z, Zhu Y, Li Y, Yu J, Zhong W, Chen Y, Lv X, Hu J, Lin J, Miao Y, Wang L. Hypoxia Induces Oxidative Injury and Apoptosis via Mediating the Nrf-2/Hippo Pathway in Blood Cells of Largemouth Bass (Micropterus salmoides). Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.841318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Investigating how aquatic animals respond to hypoxia brought about by changes in environmental temperature may be of great significance to avoid oxidative injury and maintain the quality of farmed fish in the background of global warming. Here, we investigated the effects of hypoxia on oxidative injury and environment-sensing pathway in blood cells of Micropterus salmoides. The total blood cell count (TBCC) and Giemsa staining showed that hypoxia could lead to damage of blood cells. Flow cytometry analysis confirmed that the apoptosis rate, Ca2+ level, NO production and ROS of blood cells were significantly increased under hypoxia stress. Environment-sensing pathways, such as Nrf2 pathway showed that hypoxia resulted in significant up-regulation of hiF-1 alpha subunit (Hif-1α), nuclear factor erythroid 2-related factor 2 (Nrf2) and kelch-1ike ECH- associated protein l (Keap1) expression. Meanwhile, the expression of Hippo pathway-related genes such as MOB kinase activator 1 (MOB1), large tumor suppressor homolog 1/2 (Lats1/2), yes-associated protein/transcriptional co-activator with PDZ-binding motif (YAP/TAZ), protein phosphatase 2A (PP2A) were significantly increased in blood cells after hypoxia exposure. In addition, hypoxia stress also increased the expression of catalase (CAT) and glutathione peroxidase (GPx), but decreased the expression of superoxide dismutase (SOD). Consequently, our results suggested that hypoxia could induce oxidative injury and apoptosis via mediating environment-sensing pathway such as Nrf2/Hippo pathway in blood cells of M. salmoides.
Collapse
|
11
|
Zheng XM, Yang Z, Yang GL, Huang Y, Peng JR, Wu MJ. Lung injury after cardiopulmonary bypass: Alternative treatment prospects. World J Clin Cases 2022; 10:753-761. [PMID: 35127892 PMCID: PMC8790450 DOI: 10.12998/wjcc.v10.i3.753] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/29/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Although the lung injury caused by cardiopulmonary bypass (CPB) has been extensively investigated, the incidence and mortality of lung injury after CPB remain a prominent clinical problem. The poor outcome has been attributed to multifactorial etiology, including the systemic inflammatory response and ischemia reperfusion (I/R) injury during CPB. Lung injury after CPB is a complex pathophysiological process and has many clinical manifestations of mild to severe disease. Which is associated with prognosis. To alleviate this lung injury, interventions that address the pathogenesis are particularly important. This review summarizes the pathogenesis, mechanism and treatment options of lung injury after CPB, such as lung protection with intralipid.
Collapse
Affiliation(s)
- Xue-Mei Zheng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, Sichuan Province, China
| | - Zhuo Yang
- Department of Pharmacy, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, Sichuan Province, China
| | - Guang-Li Yang
- Department of Medical Administration, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, Sichuan Province, China
| | - Yan Huang
- National Institute of Drug Clinical Trial, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, Sichuan Province, China
| | - Jie-Ru Peng
- Department of Medical Records Statistics, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610000, Sichuan Province, China
| | - Meng-Jun Wu
- Department of Anesthesiology, The Affiliated Hospital, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology, Chengdu 610000, Sichuan Province, China
| |
Collapse
|
12
|
Liu XM, Zhang Z, Zhong J, Li N, Wang T, Wang L, Zhang Q. Long non-coding RNA MALAT1 modulates myocardial ischemia-reperfusion injury though the PI3K/Akt/eNOS pathway by sponging miRNA-133a-3p to target IGF1R expression. Eur J Pharmacol 2021; 916:174719. [PMID: 34968461 DOI: 10.1016/j.ejphar.2021.174719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 01/10/2023]
Abstract
The mechanism of myocardial ischemia-reperfusion injury (MIRI) is a complex pathophysiological process that can lead to poor patient outcomes. Although LncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is reported to be highly expressed in myocardial ischemia reperfusion (IR) injury, the specific mechanism remains largely unknown. This study aimed to elucidate the roles and possible mechanism of MALAT1 in myocardial IR injury. IR model was established in rats by ligation of the anterior descending artery in vivo, and H9c2 and HL-1 cells were treated by hypoxia/reoxygenation (HR) to construct the model in vitro. The small interfering RNA (siRNA) for MALAT1 and miR-133a-3p mimics, inhibitor was used to transfect the cells. The expression of MALAT1, miR-133a-3p in MIRI were evaluated using real-time quantitative polymerase chain reaction (qRT-PCR),immunohistochemistry (IHC) and western blot (WB). Relationships between MALAT1, insulin-like growth factor 1 receptor (IGF1R) with miR-133a-3p were confirmed by luciferase reporter assay. Annexin V-FITC/PI double-labeled flow cytometry, terminal dexynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), Cell Counting Kit-8 (CCK-8), serum creatine kinase MB (CK-MB), and lactate dehydrogenase (LDH) were evaluated to examine the impact of MALAT1 on MIRI. Our results revealed that MALAT1 was highly expressed, while miR-133a-3p and IGF1R were repressed in IR and HR groups. Knockdown of MALAT1 alleviate the pro-apoptotic effect and myocardial injury in vitro and in vivo. Systematically, MALAT1 may serve as a sponge for miR-133a-3p to suppress IGF1R, which a direct target of miR-133a-3p, then inhibit the PI3K/Akt/eNOS survival pathway. Mechanistically, our study demonstrated that MALAT1 regulates PI3K/Akt/eNOS signaling via miR-133a-3p. In summary, these results suggest that MALAT1 and miR-133a-3p play important roles in MIRI. MALAT1 regulates miR-133a-3p /IGF1R axis. These results show light on the underlying mechanisms of MIRI and provide potential therapeutic targets for MIRI.
Collapse
Affiliation(s)
- Xin-Ming Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Zhenzhou Zhang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ning Li
- National Institute of Biological Sciences, Beijing, China
| | - Tao Wang
- National Institute of Biological Sciences, Beijing, China
| | - Lefeng Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Qian Zhang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
13
|
Clarke GA, Hartse BX, Niaraki Asli AE, Taghavimehr M, Hashemi N, Abbasi Shirsavar M, Montazami R, Alimoradi N, Nasirian V, Ouedraogo LJ, Hashemi NN. Advancement of Sensor Integrated Organ-on-Chip Devices. SENSORS (BASEL, SWITZERLAND) 2021; 21:1367. [PMID: 33671996 PMCID: PMC7922590 DOI: 10.3390/s21041367] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 02/06/2023]
Abstract
Organ-on-chip devices have provided the pharmaceutical and tissue engineering worlds much hope since they arrived and began to grow in sophistication. However, limitations for their applicability were soon realized as they lacked real-time monitoring and sensing capabilities. The users of these devices relied solely on endpoint analysis for the results of their tests, which created a chasm in the understanding of life between the lab the natural world. However, this gap is being bridged with sensors that are integrated into organ-on-chip devices. This review goes in-depth on different sensing methods, giving examples for various research on mechanical, electrical resistance, and bead-based sensors, and the prospects of each. Furthermore, the review covers works conducted that use specific sensors for oxygen, and various metabolites to characterize cellular behavior and response in real-time. Together, the outline of these works gives a thorough analysis of the design methodology and sophistication of the current sensor integrated organ-on-chips.
Collapse
Affiliation(s)
- Gabriel A. Clarke
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Brenna X. Hartse
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Amir Ehsan Niaraki Asli
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Mehrnoosh Taghavimehr
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Niloofar Hashemi
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran 11365, Iran;
| | - Mehran Abbasi Shirsavar
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Reza Montazami
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Nima Alimoradi
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Vahid Nasirian
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Lionel J. Ouedraogo
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Nicole N. Hashemi
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
14
|
Aydogan Kirmizi D, Baser E, Doganyigit Z. The Activation of Cannabinoid Type-2 Receptor with JWH-133 Protects Uterine Ischemia/Reperfusion-Induced Damage. Pharmacology 2020; 106:106-113. [PMID: 33105141 DOI: 10.1159/000511457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/04/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Uterus transplantation is a complex surgical procedure. Uterine ischemia/reperfusion (IR) damage occurring in this process may cause loss of function in the uterus. Cell damage must be prevented for a healthy uterine function and successful transplantation. Cannabinoids, with their increasing clinical use, are substances with strong anti-inflammatory and antioxidative effects and have a role in immune system regulation. However, their efficacy in uterine IR damage is still unknown. This study provides information on the potential applications cannabinoids agonist JWH-133 in uterine IR damage and, hence, in the transplant process. METHODS Rats were divided into 4 groups (n = 8), performed uterine IR, and treated 2 groups with JWH-133. After anesthesia, ischemia was applied for 1 h to the uterus while reperfusion was applied for 3 h. After the experiment, malondialdehyde (MDA) levels and phosphorylated nuclear factor-kappa B (p-NF-κB) expression were examined in the tissue samples. Also, cell damage was evaluated by histopathological imaging and TUNEL staining. RESULTS In the uterine IR group, NF-κB expression and MDA levels were detected at high levels. Histopathological examinations and TUNEL staining revealed extensive cell damage. On the other hand, in groups treated with JWH-133, dose-dependent NF-κB expression and MDA levels decreased (p < 0.05). Depending on the dose, the rate of surviving cells increased in TUNEL staining results. CONCLUSION The results showed that JWH-133 was effective in reducing uterine IR damage. Cannabinoids may be a new alternative that may be used in the transplantation process in the future.
Collapse
Affiliation(s)
- Demet Aydogan Kirmizi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey,
| | - Emre Baser
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Zuleyha Doganyigit
- Department of Histology and Embryology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| |
Collapse
|
15
|
Prostaglandin E1 protects cardiomyocytes against hypoxia-reperfusion induced injury via the miR-21-5p/FASLG axis. Biosci Rep 2020; 39:221373. [PMID: 31782491 PMCID: PMC6923339 DOI: 10.1042/bsr20190597] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 11/12/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Prostaglandin-E1 (PGE1) is a potent vasodilator with anti-inflammatory and antiplatelet effects. However, the mechanism by which PGE1 contributes to the amelioration of cardiac injury remains unclear. Methods: The present study was designed to investigate how PGE1 protects against hypoxia/reoxygenation (H/R)-induced injuries by regulating microRNA-21-5p (miR-21-5p) and fas ligand (FASLG). Rat H9C2 cells and isolated primary cardiomyocytes were cultured under hypoxic conditions for 6 h (6H, hypoxia for 6 h), and reoxygenated for periods of 6 (6R, reoxygenation for 6 h), 12, and 24 h, respectively. Cells from the 6H/6R group were treated with various doses of PGE1; after which, their levels of viability and apoptosis were detected. Results: The 6H/6R treatment regimen induced the maximum level of H9C2 cell apoptosis, which was accompanied by the highest levels of Bcl-2-associated X protein (Bax) and cleaved-caspase-3 expression and the lowest level of B-cell lymphoma 2 (Bcl-2) expression. Treatment with PGE1 significantly diminished the cell cytotoxicity and apoptosis induced by the 6H/6R regimen, and also decreased expression of IL-2, IL-6, P-p65, TNF-α, and cleaved-caspase-3. In addition, we proved that PGE1 up-regulated miR-21-5p expression in rat cardiomyocytes exposed to conditions that produce H/R injury. FASLG was a direct target of miR-21-5p, and PGE1 reduced the ability of H/R-injured rat cardiomyocytes to undergo apoptosis by affecting the miR-21-5p/FASLG axis. In addition, we proved that PGE1 could protect primary cardiomyocytes against H/R-induced injuries. Conclusions: These results indicate that PGE1 exerts cardioprotective effects in H9C2 cells during H/R by regulating the miR-21-5p/FASLG axis.
Collapse
|
16
|
Zajic DE, Podrabsky JE. GABA metabolism is crucial for long-term survival of anoxia in annual killifish embryos. J Exp Biol 2020; 223:jeb229716. [PMID: 32859669 DOI: 10.1242/jeb.229716] [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: 06/01/2020] [Accepted: 08/24/2020] [Indexed: 08/25/2023]
Abstract
In most vertebrates, a lack of oxygen quickly leads to irreparable damages to vital organs, such as the brain and heart. However, there are some vertebrates that have evolved mechanisms to survive periods of no oxygen (anoxia). The annual killifish (Austrofundulus limnaeus) survives in ephemeral ponds in the coastal deserts of Venezuela and their embryos have the remarkable ability to tolerate anoxia for months. When exposed to anoxia, embryos of A. limnaeus respond by producing significant amounts of γ-aminobutyric acid (GABA). This study aims to understand the role of GABA in supporting the metabolic response to anoxia. To explore this, we investigated four developmentally distinct stages of A. limnaeus embryos that vary in their anoxia tolerance. We measured GABA and lactate concentrations across development in response to anoxia and aerobic recovery. We then inhibited enzymes responsible for the production and degradation of GABA and observed GABA and lactate concentrations, as well as embryo mortality. Here, we show for the first time that GABA metabolism affects anoxia tolerance in A. limnaeus embryos. Inhibition of enzymes responsible for GABA production (glutamate decarboxylase) and degradation (GABA-transaminase and succinic acid semialdehyde dehydrogenase) led to increased mortality, supporting a role for GABA as an intermediate product and not a metabolic end-product. We propose multiple roles for GABA during anoxia and aerobic recovery in A. limnaeus embryos, serving as a neurotransmitter, an energy source, and an anti-oxidant.
Collapse
Affiliation(s)
- Daniel E Zajic
- Department of Biology, Portland State University, PO Box 751, Portland, OR 97207, USA
- Health, Human Performance, and Athletics Department, Linfield University, 900 SE Baker, McMinnville, OR 97128, USA
| | - Jason E Podrabsky
- Department of Biology, Portland State University, PO Box 751, Portland, OR 97207, USA
| |
Collapse
|
17
|
Vadlakonda L, Indracanti M, Kalangi SK, Gayatri BM, Naidu NG, Reddy ABM. The Role of Pi, Glutamine and the Essential Amino Acids in Modulating the Metabolism in Diabetes and Cancer. J Diabetes Metab Disord 2020; 19:1731-1775. [PMID: 33520860 DOI: 10.1007/s40200-020-00566-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 06/04/2020] [Indexed: 02/07/2023]
Abstract
Purpose Re-examine the current metabolic models. Methods Review of literature and gene networks. Results Insulin activates Pi uptake, glutamine metabolism to stabilise lipid membranes. Tissue turnover maintains the metabolic health. Current model of intermediary metabolism (IM) suggests glucose is the source of energy, and anaplerotic entry of fatty acids and amino acids into mitochondria increases the oxidative capacity of the TCA cycle to produce the energy (ATP). The reduced cofactors, NADH and FADH2, have different roles in regulating the oxidation of nutrients, membrane potentials and biosynthesis. Trans-hydrogenation of NADH to NADPH activates the biosynthesis. FADH2 sustains the membrane potential during the cell transformations. Glycolytic enzymes assume the non-canonical moonlighting functions, enter the nucleus to remodel the genetic programmes to affect the tissue turnover for efficient use of nutrients. Glycosylation of the CD98 (4F2HC) stabilises the nutrient transporters and regulates the entry of cysteine, glutamine and BCAA into the cells. A reciprocal relationship between the leucine and glutamine entry into cells regulates the cholesterol and fatty acid synthesis and homeostasis in cells. Insulin promotes the Pi transport from the blood to tissues, activates the mitochondrial respiratory activity, and glutamine metabolism, which activates the synthesis of cholesterol and the de novo fatty acids for reorganising and stabilising the lipid membranes for nutrient transport and signal transduction in response to fluctuations in the microenvironmental cues. Fatty acids provide the lipid metabolites, activate the second messengers and protein kinases. Insulin resistance suppresses the lipid raft formation and the mitotic slippage activates the fibrosis and slow death pathways.
Collapse
Affiliation(s)
| | - Meera Indracanti
- Institute of Biotechnology, University of Gondar, Gondar, Ethiopia
| | - Suresh K Kalangi
- Amity Stem Cell Institute, Amity University Haryana, Amity Education Valley Pachgaon, Manesar, Gurugram, HR 122413 India
| | - B Meher Gayatri
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046 India
| | - Navya G Naidu
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046 India
| | - Aramati B M Reddy
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046 India
| |
Collapse
|
18
|
Lu S, Yu L, Liu H. Trimetazidine alleviates hypoxia/reoxygenation-induced apoptosis in neonatal mice cardiomyocytes via up-regulating HMGB1 expression to promote autophagy. J Recept Signal Transduct Res 2020; 41:170-179. [PMID: 32757698 DOI: 10.1080/10799893.2020.1800736] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Previous studies demonstrated the effect of Trimetazidine (TMZ) on alleviating cardiomyocytes Hypoxia/Reoxygenation (H/R) injuries and the protective effect of autophagy on Ischemia-Reperfusion (I/R) cell injuries. However, whether the protection mechanism of TMZ was also involved in autophagy remained unclear. Our study introduces the role of HMGB1 to examine the regulation of TMZ on autophagy against cardiomyocytes H/R injuries. After cell extraction and identification through anti-α-actin staining, the cardiomyocytes were made hypoxic and reoxygenated, each for 3 h, and then treated with various concentrations of TMZ and transfected with siHMGB1. Cell viability and apoptosis were measured by the MTS method and flow cytometry, respectively. The expressions of autophagy-related factors (LC3-I, LC3-II, Beclin-1) and HMGB1 were detected by Western blot and qPCR. Lactate dehydrogenase (LDH) release was assessed by ELISA kit. The cardiomyocytes were extracted. H/R decreased the cell viability and increased the LDH level and apoptosis of cardiomyocytes. TMZ had no effect on untreated cardiomyocytes, but it reversed the adverse impact of H/R on cardiomyocytes. The expressions of LC3-II, Beclin-1, and HMGB1 and the ratio of LC3-II/LC3-I were increased in H/R-processed cardiomyocytes and further raised by TMZ pretreatment. However, siHMGB1 transfection aggravated the impact of H/R on cardiomyocytes and suppressed the protective effects of TMZ on H/R damaged cardiomyocytes by increasing the LDH level and apoptosis and reducing the viability of cardiomyocytes. Autophagy was inhibited by siHMGB1 in TMZ-pretreated and H/R-processed cardiomyocytes. TMZ protected cardiomyocytes against H/R injuries may through regulating HMGB1 to increase the impact of autophagy.
Collapse
Affiliation(s)
- Shiwen Lu
- Department of Cardiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lifei Yu
- Department of Cardiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hao Liu
- Department of Cardiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
19
|
Sun Y, Dong H, Zhan A, Wang W, Duan Y, Xie M, Liu Q, Li H, Zhang J. Protection of teprenone against hypoxia and reoxygenation stress in stomach and intestine of Lateolabrax maculatus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:575-584. [PMID: 31900796 DOI: 10.1007/s10695-019-00732-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 11/05/2019] [Indexed: 06/10/2023]
Abstract
Teprenone (geranylgeranylacetone) is one kind of safe and effective agent in gastrointestinal mucosa, which have been widely used in human and veterinary, but rarely used in aquaculture animals. In this study, Lateolabrax maculatus, an important economic fish species in southern China, was taken as the object of study to investigate the protective effect of teprenone on intestinal stress. The present study was designed to investigate the potential mechanism underlying the protection offered by teprenone to protect the gastrointestinal tract against hypoxia and reoxygenation injury of L. maculatus. (a) For oxidative stress parameters, SOD, CAT, and T-AOC in control group were higher than those in teprenone group. MDA content was significantly higher than that in teprenone group at N and 12h time points in intestine (P < 0.05), and at 12, 24, and 48 h time points in stomach. (b) For immune-associated proteins, LZM activity in the control group was lower than that in the teprenone group, and the difference between the two groups in stomach and intestine was significant at 12.48 h and 6.48 h time points, respectively (P < 0.05). Compared with time point N, the content of HSP70 in the control group increased at 0 h in intestine. At 0-48 h, intestine HSP70 content in the control group showed a gradually decreasing trend, which was higher than that in the teprenone group. (c) For apoptosis-related factors, the activity of Cyt-C, caspase9, and caspase3 increased first and then decreased in both groups. The content of Cyt-C in the control group was significantly higher than that in the teprenone group at N-3.6 h, and 3.48 h time points in stomach and intestine, respectively (P<0.05). The activity of caspase9 and caspase3 was higher than that in the teprenone group at N-48 h. Results indicated that acute hypoxia and reoxygenation cause the expression levels of oxidative stress and apoptosis-related factors in the stomach and intestine increased first and then decreased within 0-48 h. Acute hypoxia and reoxygenation also that causes the level of nonspecific immunity decreased first and then increased. A total of 400-mg/kg treatment of teprenone can protect stomach and intestinal tissues to a certain extent. It can effectively protect oxidative stress and apoptosis within 0-48 h after acute hypoxia and reoxygenation and enhance non-specific immunity.
Collapse
Affiliation(s)
- YongXu Sun
- Key Lab. of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - HongBiao Dong
- Key Lab. of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
| | - AiJun Zhan
- Shenzhen Academy of Inspection and Quarantine, Shenzhen, 518001, China
| | - WenHao Wang
- Key Lab. of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - YaFei Duan
- Key Lab. of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
| | - Mujiao Xie
- Key Lab. of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
| | - QingSong Liu
- Key Lab. of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
| | - Hua Li
- Key Lab. of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China
| | - JiaSong Zhang
- Key Lab. of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, China.
- Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen, 518121, China.
| |
Collapse
|
20
|
Allen KN, Vázquez-Medina JP. Natural Tolerance to Ischemia and Hypoxemia in Diving Mammals: A Review. Front Physiol 2019; 10:1199. [PMID: 31620019 PMCID: PMC6763568 DOI: 10.3389/fphys.2019.01199] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
Reperfusion injury follows ischemia/reperfusion events occurring during myocardial infarction, stroke, embolism, and other peripheral vascular diseases. Decreased blood flow and reduced oxygen tension during ischemic episodes activate cellular pathways that upregulate pro-inflammatory signaling and promote oxidant generation. Reperfusion after ischemia recruits inflammatory cells to the vascular wall, further exacerbating oxidant production and ultimately resulting in cell death, tissue injury, and organ dysfunction. Diving mammals tolerate repetitive episodes of peripheral ischemia/reperfusion as part of the cardiovascular adjustments supporting long duration dives. These adjustments allow marine mammals to optimize the use of their body oxygen stores while diving but can result in selectively reduced perfusion to peripheral tissues. Remarkably, diving mammals show no apparent detrimental effects associated with these ischemia/reperfusion events. Here, we review the current knowledge regarding the strategies marine mammals use to suppress inflammation and cope with oxidant generation potentially derived from diving-induced ischemia/reperfusion.
Collapse
|
21
|
Kakaroubas N, Brennan S, Keon M, Saksena NK. Pathomechanisms of Blood-Brain Barrier Disruption in ALS. NEUROSCIENCE JOURNAL 2019; 2019:2537698. [PMID: 31380411 PMCID: PMC6652091 DOI: 10.1155/2019/2537698] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/18/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier (BBB) and the blood-spinal cord barrier (BSCB) are responsible for controlling the microenvironment within neural tissues in humans. These barriers are fundamental to all neurological processes as they provide the extreme nutritional demands of neural tissue, remove wastes, and maintain immune privileged status. Being a semipermeable membrane, both the BBB and BSCB allow the diffusion of certain molecules, whilst restricting others. In amyotrophic lateral sclerosis (ALS) and other neurodegenerative diseases, these barriers become hyperpermeable, allowing a wider variety of molecules to pass through leading to more severe and more rapidly progressing disease. The intention of this review is to discuss evidence that BBB hyperpermeability is potentially a disease driving feature in ALS and other neurodegenerative diseases. The various biochemical, physiological, and genomic factors that can influence BBB permeability in ALS and other neurodegenerative diseases are also discussed, in addition to novel therapeutic strategies centred upon the BBB.
Collapse
Affiliation(s)
- Nicholas Kakaroubas
- Neurodegenerative Disease Section, Iggy Get Out, 19A Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (University of NSW), Chancellery Walk, Kensington NSW 2033, Sydney, Australia
| | - Samuel Brennan
- Neurodegenerative Disease Section, Iggy Get Out, 19A Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Matthew Keon
- Neurodegenerative Disease Section, Iggy Get Out, 19A Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Nitin K. Saksena
- Neurodegenerative Disease Section, Iggy Get Out, 19A Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| |
Collapse
|
22
|
Spinal cord injury: pathophysiology, treatment strategies, associated challenges, and future implications. Cell Tissue Res 2019; 377:125-151. [PMID: 31065801 DOI: 10.1007/s00441-019-03039-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Abstract
Axonal regeneration and formation of tripartite (axo-glial) junctions at damaged sites is a prerequisite for early repair of injured spinal cord. Transplantation of stem cells at such sites of damage which can generate both neuronal and glial population has gained impact in terms of recuperation upon infliction with spinal cord injury. In spite of the fact that a copious number of pre-clinical studies using different stem/progenitor cells have shown promising results at acute and subacute stages, at the chronic stages of injury their recovery rates have shown a drastic decline. Therefore, developing novel therapeutic strategies are the need of the hour in order to assuage secondary morbidity and effectuate improvement of the spinal cord injury (SCI)-afflicted patients' quality of life. The present review aims at providing an overview of the current treatment strategies and also gives an insight into the potential cell-based therapies for the treatment of SCI.
Collapse
|
23
|
Renoprotective Effect of the Histone Deacetylase Inhibitor CG200745 in DOCA-Salt Hypertensive Rats. Int J Mol Sci 2019; 20:ijms20030508. [PMID: 30691015 PMCID: PMC6387176 DOI: 10.3390/ijms20030508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
The novel histone deacetylase inhibitor CG200745 was initially developed to treat various hematological and solid cancers. We investigated the molecular mechanisms associated with the renoprotective effects of CG200745 using deoxycorticosterone acetate (DOCA)-salt hypertensive (DSH) rats. DOCA strips (200 mg/kg) were implanted into rats one week after unilateral nephrectomy. Two weeks after DOCA implantation, DSH rats were randomly divided into two groups that received either physiological saline or CG200745 (5 mg/kg/day) for another two weeks. The extent of glomerulosclerosis and tubulointerstitial fibrosis was determined by Masson's trichrome staining. The renal expression of fibrosis and inflammatory markers was detected by semiquantitative immunoblotting, a polymerase chain reaction, and immunohistochemistry. Pathological signs such as glomerulosclerosis, tubulointerstitial fibrosis, increased systolic blood pressure, decreased creatinine clearance, and increased albumin-to-creatinine ratios in DSH rats were alleviated by CG200745 treatment compared to those manifestations in positive control animals. Furthermore, this treatment counteracted the increased expression of αSMA, TGF-β1, and Bax, and the decreased expression of Bcl-2 in the kidneys of DSH rats. It also attenuated the increase in the number of apoptotic cells in DSH rats. Thus, CG200745 can effectively prevent the progression of renal injury in DSH rats by exerting anti-inflammatory, anti-fibrotic, and anti-apoptotic effects.
Collapse
|
24
|
Lu WJ, Lin KH, Tseng MF, Yuan KC, Huang HC, Sheu JR, Chen RJ. New therapeutic strategy of hinokitiol in haemorrhagic shock-induced liver injury. J Cell Mol Med 2018; 23:1723-1734. [PMID: 30548082 PMCID: PMC6378182 DOI: 10.1111/jcmm.14070] [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: 08/09/2018] [Revised: 11/04/2018] [Accepted: 11/12/2018] [Indexed: 01/19/2023] Open
Abstract
Haemorrhagic shock and resuscitation (HS/R) may cause global ischaemia-reperfusion injury, which can result in systemic inflammation, multiorgan failure (particularly liver failure) and high mortality. Hinokitiol, a bioactive tropolone-related compound, exhibits antiplatelet and anti-inflammatory activities. Targeting inflammatory responses is a potential strategy for ameliorating hepatic injury during HS/R. Whether hinokitiol prevents hepatic injury during HS/R remains unclear. In the present study, we determined the role of hinokitiol following HS/R. The in vivo assays revealed that hinokitiol markedly attenuated HS/R-induced hepatic injury. Hinokitiol could inhibited NF-κB activation and IL-6 and TNF-α upregulation in liver tissues. Moreover, hinokitiol reduced caspase-3 activation, upregulated Bax and downregulated Bcl-2. These findings suggest that hinokitiol can ameliorate liver injury following HS/R, partly through suppression of inflammation and apoptosis. Furthermore, the in vitro data revealed that hinokitiol significantly reversed hypoxia/reoxygenation (H/R)-induced cell death and apoptosis in the primary hepatocytes. Hinokitiol prevented H/R-induced caspase-3 activation, PPAR cleavage, Bax overexpression and Bcl-2 downregulation. Moreover, hinokitiol attenuated H/R-stimulated NF-κB activation and reduced the levels of IL-6 and TNF-α mRNAs, suggesting that hinokitiol can protect hepatocytes from H/R injury. Collectively, our data suggest that hinokitiol attenuates liver injury following HS/R, partly through the inhibition of NF-κB activation.
Collapse
Affiliation(s)
- Wan-Jung Lu
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Metabolism and Obesity Sciences, College of Public Health and Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Hung Lin
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Mei-Fang Tseng
- Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Ching Yuan
- Department of Emergency and Critical Care Medicine and Division of Acute Care Surgery and Trauma, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hung-Chang Huang
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Joen-Rong Sheu
- Graduate Institute of Metabolism and Obesity Sciences, College of Public Health and Nutrition, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Ray-Jade Chen
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
25
|
Zhang RY, Qiao ZY, Liu HJ, Ma JW. Sonic hedgehog signaling regulates hypoxia/reoxygenation-induced H9C2 myocardial cell apoptosis. Exp Ther Med 2018; 16:4193-4200. [PMID: 30344694 DOI: 10.3892/etm.2018.6678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 06/29/2018] [Indexed: 12/12/2022] Open
Abstract
The sonic hedgehog (Shh) signaling pathway has been reported to protect cells against hypoxia/reoxygenation (H/R) injury; however, the role of Shh and relevant molecular mechanisms remain unclear. In the present study, the rat cardiomyoblast cell line H9C2 was subjected to hypoxia and serum-starvation for 4 h. Cells were subsequently reoxygenated using 95% O2 and 5% CO2. Reverse transcription-quantitative polymerase chain reaction was performed to quantify the expression of Shh mRNA, while cell apoptosis was assessed using flow cytometry. Caspase-3 activity and p53 expression were measured by western blotting and an MTT assay was subsequently used to assess cell viability. In addition, reactive oxygen species levels were measured using dichlorofluorescein and H/R-induced changes in the activation of superoxide dismutase, catalase, phosphorylated-endothelial nitric oxide synthase, phosphorylated-protein kinase B (Akt) and mammalian target of rapamycin activation were assessed using western blotting. H/R treatment decreased the cell viability of H9C2 cells, but activated endogenous Shh signaling. The activation of Shh signaling protected H9C2 myocardial cells from H/R-induced apoptosis and restored cell viability. In the present study, Shh signaling was demonstrated to serve a protective role against H/R by activating the phosphoinositol 3-kinase (PI3K)/Akt pathway and promoting the expression of anti-oxidant enzymes to ameliorate oxidative stress. In summary, Shh signaling attenuated H/R-induced apoptosis through via the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Rui-Ying Zhang
- Department of Cardiology, Fengxian District Central Hospital, Shanghai 201400, P.R. China
| | - Zeng-Yong Qiao
- Department of Cardiology, Fengxian District Central Hospital, Shanghai 201400, P.R. China
| | - Hua-Jin Liu
- Department of Cardiology, Fengxian District Central Hospital, Shanghai 201400, P.R. China
| | - Jiang-Wei Ma
- Department of Cardiology, Fengxian District Central Hospital, Shanghai 201400, P.R. China
| |
Collapse
|
26
|
Ietta F, Ferro EAV, Bevilacqua E, Benincasa L, Maioli E, Paulesu L. Role of the Macrophage Migration Inhibitory Factor (MIF) in the survival of first trimester human placenta under induced stress conditions. Sci Rep 2018; 8:12150. [PMID: 30108299 PMCID: PMC6092320 DOI: 10.1038/s41598-018-29797-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/10/2018] [Indexed: 11/09/2022] Open
Abstract
Macrophage Migration Inhibitory Factor (MIF) is a multifunctional molecule highly secreted by human placenta mainly in the early phases of pregnancy. Studies in different cells show that MIF is a pro-survival factor by binding to its receptor CD74. By using the in vitro model of placental explants from first trimester pregnancy, we investigated the role of MIF in the survival of placental cells under induced stress conditions that promote apoptosis or mimic the hypoxia/re-oxygenation (H/R) injury that placenta could suffer in vivo. We demonstrated that recombinant MIF (rMIF) treatment was able to reduce caspase-3 activation when cultures were challenged with the apoptosis-inducer Carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP) while, in the cultures exposed to H/R, the treatment with rMIF did not show any effect. However, a significant increase in caspase-3 and caspase-8 activation was found when H/R-exposed cultures, were treated with anti-MIF or anti-CD74 antibody. We also observed that under H/R, a significant amount of endogenous MIF was released into the medium, which could account for the lack of effect of rMIF added to the cultures. Our results demonstrate for the first time that the MIF/CD74 axis contributes to maintain trophoblast homeostasis, by preventing abnormal apoptotic death.
Collapse
Affiliation(s)
- Francesca Ietta
- Department of Life Sciences, University of Siena, Via A. Moro 4, 53100, Siena, Italy.
| | - Eloisa Amália Vieira Ferro
- Laboratory of Immunophysiology of Reproduction, Institute of Biomedical Sciences, Federal University of Uberlândia, Av. Pará 1720, 38405320, Uberlândia, Brazil
| | - Estela Bevilacqua
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof Lineu Prestes 1524, 05508-900, São Paulo, Brazil
| | - Linda Benincasa
- Department of Life Sciences, University of Siena, Via A. Moro 4, 53100, Siena, Italy
| | - Emanuela Maioli
- Department of Life Sciences, University of Siena, Via A. Moro 4, 53100, Siena, Italy
| | - Luana Paulesu
- Department of Life Sciences, University of Siena, Via A. Moro 4, 53100, Siena, Italy
| |
Collapse
|
27
|
Li C, Wang K, Guo L, Sun H, Huang H, Lin X, Li Q. Inhibition of miR-34a-5p alleviates hypoxia-reoxygenation injury by enhancing autophagy in steatotic hepatocytes. Biol Open 2018; 7:7/3/bio033290. [PMID: 29581146 PMCID: PMC5898271 DOI: 10.1242/bio.033290] [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] [Indexed: 12/20/2022] Open
Abstract
Hypoxia-reoxygenation (H/R) injury in steatotic hepatocytes has been implicated in liver dysfunction after liver transplantation. MicroRNAs (miRs) play important roles in regulating several cell biology mechanisms related to H/R injury. However, the role of miRs in regulating H/R injury in steatotic hepatocytes is still unclear. We established an in vitro model for studying H/R injury in steatotic hepatocytes and identified miR-34a-5p as a miR that was substantially upregulated in steatotic hepatocytes under H/R challenge. MiR-34a-5p expression was modified by transfecting miR-34a-5p mimic and inhibitor into H/R-challenged steatotic hepatocytes. We found that inhibition of miR-34a-5p alleviated H/R-induced apoptosis and promoted post-H/R proliferation in steatotic hepatocytes. Whereas, overexpression of miR-34a-5p augmented H/R-induced apoptosis and prohibited post-H/R proliferation. By examining autophagy, our data demonstrated that miR-34a-5p suppressed autophagy in H/R-challenged steatotic hepatocytes, induction of autophagy partially rescued the exaggeration of H/R injury induced by miR-34a-5p mimic, while inhibition of autophagy impaired the protection of the miR-34a-5p inhibitor against H/R injury. In conclusion, miR-34a-5p is crucial in exaggerating H/R injury, likely by suppressing autophagy in steatotic hepatocytes. Inhibition of miR-34a may be a promising strategy to protect steatotic hepatocytes against H/R-injury.
Collapse
Affiliation(s)
- Chuanjiang Li
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China .,Department of Surgery, Linzhi Municipal People's Hospital, Linzhi, Tibet 860100, China
| | - Kai Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Linghong Guo
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hang Sun
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hai Huang
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | - XinXin Lin
- The First Clinical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qingping Li
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
28
|
Baust JM, Corwin W, Snyder KK, Van Buskirk R, Baust JG. Cryopreservation: Evolution of Molecular Based Strategies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 951:13-29. [PMID: 27837551 DOI: 10.1007/978-3-319-45457-3_2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cryopreservation (CP) is an enabling process providing for on-demand access to biological material (cells and tissues) which serve as a starting, intermediate or even final product. While a critical tool, CP protocols, approaches and technologies have evolved little over the last several decades. A lack of conversion of discoveries from the CP sciences into mainstream utilization has resulted in a bottleneck in technological progression in areas such as stem cell research and cell therapy. While the adoption has been slow, discoveries including molecular control and buffering of cell stress response to CP as well as the development of new devices for improved sample freezing and thawing are providing for improved CP from both the processing and sample quality perspectives. Numerous studies have described the impact, mechanisms and points of control of cryopreservation-induced delayed-onset cell death (CIDOCD). In an effort to limit CIDOCD, efforts have focused on CP agent and freeze media formulation to provide a solution path and have yielded improvements in survival over traditional approaches. Importantly, each of these areas, new technologies and cell stress modulation, both individually and in combination, are now providing a new foundation to accelerate new research, technology and product development for which CP serves as an integral component. This chapter provides an overview of the molecular stress responses of cells to cryopreservation, the impact of the hypothermic and cell death continuums and the targeted modulation of common and/or cell specific responses to CP in providing a path to improving cell quality.
Collapse
Affiliation(s)
- John M Baust
- CPSI Biotech, 2 Court St, Owego, NY, 13827, USA. .,Institute of Biomedical Technology, State University of New York at Binghamton, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA.
| | - William Corwin
- Department of Immunology and Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Kristi K Snyder
- CPSI Biotech, 2 Court St, Owego, NY, 13827, USA.,Institute of Biomedical Technology, State University of New York at Binghamton, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Robert Van Buskirk
- CPSI Biotech, 2 Court St, Owego, NY, 13827, USA.,Institute of Biomedical Technology, State University of New York at Binghamton, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA.,Department of Biological Sciences, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - John G Baust
- Institute of Biomedical Technology, State University of New York at Binghamton, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA.,Department of Biological Sciences, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| |
Collapse
|
29
|
Reed DK, Carter A, Dixit M, Arany I. p66shc-mediated toxicity of high-dose α-tocopherol in renal proximal tubule cells. J Physiol Biochem 2017; 73:267-273. [DOI: 10.1007/s13105-017-0551-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/18/2017] [Indexed: 10/20/2022]
|
30
|
Sahin Ersoy G, Kurek Eken M, Cevik O, Cilingir OT, Tal R. Mycophenolate mofetil attenuates uterine ischaemia/reperfusion injury in a rat model. Reprod Biomed Online 2016; 34:115-123. [PMID: 27913135 DOI: 10.1016/j.rbmo.2016.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 11/03/2016] [Accepted: 11/08/2016] [Indexed: 10/20/2022]
Abstract
This study evaluated the effect of mycophenolate mofetil (MMF) on uterine tissue preservation following ischaemia/reperfusion (I/R) injury. Uterine I/R injury was induced in rats by clamping the lower abdominal aorta and ovarian arteries for 30 min. Group I/R + V (n = 7) received vehicle alone while Group I/R + M (n = 7) received 20 mg/kg/day MMF. Control groups underwent sham surgery and received vehicle (Group C) or 20 mg/kg/day MMF (Group M) (n = 7 for both). Four hours after detorsion, uterine tissue 8-hydroxy-2'-deoxyguanosine (8-OHdG), glutathione, malondialdehyde (MDA), myeloperoxidase (MPO), superoxide dismutase (SOD) and serum ischaemia modified albumin (IMA) concentrations were measured. Histopathological analyses were performed. The I/R + M group showed significant reduction in serum IMA and uterine tissue 8-OHdG, MDA and MPO and significant increase in SOD concentrations compared with the I/R + V group, indicating a protective effect against I/R oxidative damage (P = 0.009, P = 0.006, P = 0.002, P = 0.003 and P = 0.009, respectively). Histopathological evaluation revealed MMF treatment resulted in significantly less tissue and cellular damage and apoptosis compared with the I/R + V group. These results indicate MMF is effective in attenuating uterine tissue damage and preventing apoptosis following uterine I/R injury, probably via anti-inflammatory and anti-oxidative action.
Collapse
Affiliation(s)
- Gulcin Sahin Ersoy
- Department of Obstetrics and Gynecology, Kartal Dr Lutfi Kirdar Education and Research Hospital, Istanbul, Turkey.
| | - Meryem Kurek Eken
- Department of Obstetrics and Gynecology, Zeynep Kamil Education and Research Hospital, Istanbul, Turkey
| | - Ozge Cevik
- Department of Biochemistry, Faculty of Pharmacy, Cumhuriyet University, Sivas, Turkey
| | - Ozlem T Cilingir
- Department of Histology and Embryology, Marmara University School of Medicine, Istanbul, Turkey
| | - Reshef Tal
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
31
|
Ischémie–reperfusion. Liquides de conservation et machines de perfusion en transplantation rénale. Prog Urol 2016; 26:964-976. [DOI: 10.1016/j.purol.2016.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022]
|
32
|
Cao H, Feng Y, Ning Y, Zhang Z, Li W, Li Q. Edaravone protects rats and human pulmonary alveolar epithelial cells against hyperoxia injury: heme oxygenase-1 and PI3K/Akt pathway may be involved. Exp Lung Res 2016; 41:404-14. [PMID: 26151294 DOI: 10.3109/01902148.2015.1054053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE/AIM Hyperoxic acute lung injury (HALI) is a clinical syndrome as a result of prolonged supplement of high concentrations of oxygen. As yet, no specific treatment is available for HALI. The present study aims to investigate the effects of edaravone on hyperoxia-induced oxidative injury and the underlying mechanism. MATERIALS AND METHODS We treated rats and human pulmonary alveolar epithelial cells with hyperoxia and different concentration of edaravone, then examined the effects of edaravone on cell viability, cell injury and two oxidative products. The roles of heme oxygenase-1 (HO-1) and PI3K/Akt pathway were explored using Western blot and corresponding inhibitors. RESULTS The results showed that edaravone reduced lung biochemical alterations induced by hyperoxia and mortality of rats, dose-dependently alleviated cell mortality, cell injury, and peroxidation of cellular lipid and DNA oxidative damage. It upregulated cellular HO-1 expression and activity, which was reversed by PI3K/Akt pathway inhibition. The administration of zinc protoporphyrin-IX, a HO-1 inhibitor, and LY249002, a PI3K/Akt pathway inhibitor, abolished the protective effects of edaravone in cells. CONCLUSIONS This study indicates that edaravone protects rats and human pulmonary alveolar epithelial cells against hyperoxia-induced injury and the antioxidant effect may be related to upregulation of HO-1, which is regulated by PI3K/Akt pathway.
Collapse
Affiliation(s)
- Huifang Cao
- a 1 Department of Respiratory Diseases, ChangHai Hospital , Second Military Medical University , Shanghai, China
| | - Ying Feng
- b 2 Department of Respiratory Diseases, Jing'an District Centre Hospital of Shanghai , Huashan Hospital Fudan University, Jing'An Branch , Shanghai, China
| | - Yunye Ning
- a 1 Department of Respiratory Diseases, ChangHai Hospital , Second Military Medical University , Shanghai, China
| | - Zinan Zhang
- b 2 Department of Respiratory Diseases, Jing'an District Centre Hospital of Shanghai , Huashan Hospital Fudan University, Jing'An Branch , Shanghai, China
| | - Weihao Li
- b 2 Department of Respiratory Diseases, Jing'an District Centre Hospital of Shanghai , Huashan Hospital Fudan University, Jing'An Branch , Shanghai, China
| | - Qiang Li
- a 1 Department of Respiratory Diseases, ChangHai Hospital , Second Military Medical University , Shanghai, China
| |
Collapse
|
33
|
Abdul-Muneer PM, Long M, Conte AA, Santhakumar V, Pfister BJ. High Ca 2+ Influx During Traumatic Brain Injury Leads to Caspase-1-Dependent Neuroinflammation and Cell Death. Mol Neurobiol 2016; 54:3964-3975. [PMID: 27289225 DOI: 10.1007/s12035-016-9949-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/06/2016] [Indexed: 12/22/2022]
Abstract
We investigated the hypothesis that high Ca2+ influx during traumatic brain injury induces the activation of the caspase-1 enzyme, which triggers neuroinflammation and cell apoptosis in a cell culture model of neuronal stretch injury and an in vivo model of fluid percussion injury (FPI). We first established that stretch injury causes a rapid increase in the intracellular Ca2+ level, which activates interleukin-converting enzyme caspase-1. The increase in the intracellular Ca2+ level and subsequent caspase-1 activation culminates into neuroinflammation via the maturation of IL-1β. Further, we analyzed caspase-1-mediated apoptosis by TUNEL staining and PARP western blotting. The voltage-gated sodium channel blocker, tetrodotoxin, mitigated the stretch injury-induced neuroinflammation and subsequent apoptosis by blocking Ca2+ influx during the injury. The effect of tetrodotoxin was similar to the caspase-1 inhibitor, zYVAD-fmk, in neuronal culture. To validate the in vitro results, we demonstrated an increase in caspase-1 activity, neuroinflammation and neurodegeneration in fluid percussion-injured animals. Our data suggest that neuronal injury/traumatic brain injury (TBI) can induce a high influx of Ca2+ to the cells that cause neuroinflammation and cell death by activating caspase-1, IL-1β, and intrinsic apoptotic pathways. We conclude that excess IL-1β production and cell death may contribute to neuronal dysfunction and cognitive impairment associated with TBI.
Collapse
Affiliation(s)
- P M Abdul-Muneer
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA. .,Neuroscience Institute, JFK Medical Center, Edison, NJ, 08820, USA.
| | - Mathew Long
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Adriano Andrea Conte
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Bryan J Pfister
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
| |
Collapse
|
34
|
Weinberg JM, Bienholz A, Venkatachalam MA. The role of glycine in regulated cell death. Cell Mol Life Sci 2016; 73:2285-308. [PMID: 27066896 PMCID: PMC4955867 DOI: 10.1007/s00018-016-2201-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 01/22/2023]
Abstract
The cytoprotective effects of glycine against cell death have been recognized for over 28 years. They are expressed in multiple cell types and injury settings that lead to necrosis, but are still not widely appreciated or considered in the conceptualization of cell death pathways. In this paper, we review the available data on the expression of this phenomenon, its relationship to major pathophysiologic pathways that lead to cell death and immunomodulatory effects, the hypothesis that it involves suppression by glycine of the development of a hydrophilic death channel of molecular dimensions in the plasma membrane, and evidence for its impact on disease processes in vivo.
Collapse
Affiliation(s)
- Joel M Weinberg
- Division of Nephrology, Department of Internal Medicine, Veterans Affairs Ann Arbor Healthcare System and University of Michigan, Room 1560, MSRB II, Ann Arbor, MI, 48109-0676, USA.
| | - Anja Bienholz
- Department of Nephrology, University Duisburg-Essen, 45122, Essen, Germany
| | - M A Venkatachalam
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX, 78234, USA
| |
Collapse
|
35
|
Alnasser HA, Guan Q, Zhang F, Gleave ME, Nguan CYC, Du C. Requirement of clusterin expression for prosurvival autophagy in hypoxic kidney tubular epithelial cells. Am J Physiol Renal Physiol 2016; 310:F160-73. [DOI: 10.1152/ajprenal.00304.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/05/2015] [Indexed: 02/08/2023] Open
Abstract
Cellular autophagy is a prosurvival mechanism in the kidney against ischemia-reperfusion injury (IRI), but the molecular pathways that activate the autophagy in ischemic kidneys are not fully understood. Clusterin (CLU) is a chaperone-like protein, and its expression is associated with kidney resistance to IRI. The present study investigated the role of CLU in prosurvival autophagy in the kidney. Renal IRI was induced in mice by clamping renal pedicles at 32°C for 45 min. Hypoxia in renal tubular epithelial cell (TEC) cultures was induced by exposure to a 1% O2 atmosphere. Autophagy was determined by either light chain 3-BII expression with Western blot analysis or light chain 3-green fluorescent protein aggregation with confocal microscopy. Cell apoptosis was determined by flow cytometric analysis. The unfolded protein response was determined by PCR array. Here, we showed that autophagy was significantly activated by IRI in wild-type (WT) but not CLU-deficient kidneys. Similarly, autophagy was activated by hypoxia in human proximal TECs (HKC-8) and WT mouse primary TECs but was impaired in CLU-null TECs. Hypoxia-activated autophagy was CLU dependent and positively correlated with cell survival, and inhibition of autophagy significantly promoted cell death in both HKC-8 and mouse WT/CLU-expressing TECs but not in CLU-null TECs. Further experiments showed that CLU-dependent prosurvival autophagy was associated with activation of the unfolded protein response in hypoxic kidney cells. In conclusion, these data suggest that activation of prosurvival autophagy by hypoxia in kidney cells requires CLU expression and may be a key cytoprotective mechanism of CLU in the protection of the kidney from hypoxia/ischemia-mediated injury.
Collapse
Affiliation(s)
- Hatem A. Alnasser
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Qiunong Guan
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; and
| | - Fan Zhang
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Martin E. Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Christopher Y. C. Nguan
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Caigan Du
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; and
| |
Collapse
|
36
|
Zhao P, Li F, Gao W, Wang J, Fu L, Chen Y, Huang M. Angiotensin1-7 protects cardiomyocytes from hypoxia/reoxygenation-induced oxidative stress by preventing ROS-associated mitochondrial dysfunction and activating the Akt signaling pathway. Acta Histochem 2015; 117:803-10. [PMID: 26251197 DOI: 10.1016/j.acthis.2015.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/17/2015] [Accepted: 07/24/2015] [Indexed: 01/12/2023]
Abstract
Angiotensin1-7 (Ang1-7) is a biologically active member of the renin-angiotensin system, which has been reported to exhibit protective effect in myocardial ischemia reperfusion-induced injury. However, the molecular basis of this effect is not well understood. It has been proposed that oxidative stress-induced cardiomyocyte apoptosis is a major consequence of hypoxia/reoxygenation (H/R) injury. This study investigates the protective effect of Ang1-7 against H/R-induced oxidative stress in rat H9C2 cells. Our results showed that Ang1-7 (80nM) treatment significantly protected cells from H/R-induced oxidative injury via improving cell viability and reducing cell apoptosis. The protective effect of Ang1-7 was associated with the inhibition of ROS-associated mitochondrial dysfunction as well as the induction of Akt phosphorylation. These findings may significantly contribute to better understanding the protective effect of Ang1-7, particularly in hypoxia/reoxygenation-induced heart diseases and form the basis in the therapeutic development in treating cardiovascular diseases.
Collapse
Affiliation(s)
- Pengjun Zhao
- Department of Cardiology, Children's Medical Center Affiliated to Shanghai Jiao Tong University Medical College, Shanghai 200127, China
| | - Fen Li
- Department of Cardiology, Children's Medical Center Affiliated to Shanghai Jiao Tong University Medical College, Shanghai 200127, China.
| | - Wei Gao
- Department of Cardiology, Children's Medical Center Affiliated to Shanghai Jiao Tong University Medical College, Shanghai 200127, China
| | - Jing Wang
- Department of Cardiology, Children's Medical Center Affiliated to Shanghai Jiao Tong University Medical College, Shanghai 200127, China
| | - Lijun Fu
- Department of Cardiology, Children's Medical Center Affiliated to Shanghai Jiao Tong University Medical College, Shanghai 200127, China
| | - Yiwei Chen
- Department of Cardiology, Children's Medical Center Affiliated to Shanghai Jiao Tong University Medical College, Shanghai 200127, China
| | - Meirong Huang
- Department of Cardiology, Children's Medical Center Affiliated to Shanghai Jiao Tong University Medical College, Shanghai 200127, China
| |
Collapse
|
37
|
Zhang Q, Wang G, Yuan W, Wu J, Wang M, Li C. The effects of phosphodiesterase-5 inhibitor sildenafil against post-resuscitation myocardial and intestinal microcirculatory dysfunction by attenuating apoptosis and regulating microRNAs expression: essential role of nitric oxide syntheses signaling. J Transl Med 2015; 13:177. [PMID: 26040988 PMCID: PMC4467614 DOI: 10.1186/s12967-015-0550-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/26/2015] [Indexed: 02/06/2023] Open
Abstract
Background Recent experimental and clinical studies have indicated the cardioprotective role of sildenafil during ischemia/reperfusion (I/R) injury. Sildenafil has been shown to attenuate postresuscitation myocardial dysfunction in piget models of ventricular fibrillation. This study was designed to investigate if administration of sildenafil will attenuate post-resuscitation myocardial dysfunction by attenuating apoptosis and regulating miRNA expressions, furthermore, ameliorating the severity of post-microcirculatory dysfunction. Methods Twenty-four male pigs (weighing 30 ± 2 kg) were randomly divided into groups, sildenafil pretreatment (n = 8), saline (n = 8) and sham operation (sham, n = 8). Sildenafil pretreatment consisted of 0.5 mg/kg sildenafil, administered once intraperitoneally 30 min prior to ventricular fibrillation (VF). Eight minutes of untreated VF was followed by defibrillation in anesthetized, closed-chest pigs. Hemodynamic status and blood samples were obtained at 0 min, 0.5, 1, 2, 4 and 6 h after return of spontaneous circulation (ROSC). Surviving pigs were euthanatized at 24 h after ROSC, and hearts were removed for analysis by electron microscopy, western blotting, quantitative real-time polymerase chain reaction (PCR), and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. Intestinal microcirculatory blood flow was visualized by a sidestream dark-field imaging device at baseline and 0.5, 1, 2, 4, and 6 h after ROSC. Results Compared with the saline group, the sildenafil group had a higher 24-hour survival (7/8 versus 3/8 survivors, p < 0.05) and a better outcome in hemodynamic parameters. The protective effect of sildenafil also correlated with reduced cardiomyocyte apoptosis, as evidenced by reduced TUNEL-positive cells, increased anti-apoptotic Bcl-2/Bax ratio and inhibited caspase-3 activity in myocardium. Additionally, sildenafil treatment inhibited the increases in the microRNA-1 levels and alleviated the decreases in the microRNA-133a levels which negatively regulates pro-apoptotic genes. At 6 h after ROSC, post-resuscitation perfused vessel density and microcirculatory flow index were significantly lower in the saline group than in the sildenafil group. Conclusions The major findings of this study are as follows: (1) sildenafil improved post-resuscitation perfusion of the heart, and thus reduced cardiac myocyte apoptosis and improved cardiac function; (2) sildenafil treatment inhibited the increases in the microRNA-1 levels, but alleviated the decreases in the microRNA-133a levels.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Guoxing Wang
- Department of Emergency Medicine, Beijing You-yi Hospital, Capital Medical University, Beijing, 100050, China.
| | - Wei Yuan
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Junyuan Wu
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Miaomiao Wang
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - ChunSheng Li
- Department of Emergency Medicine, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
38
|
Cohen A, Lerner-Yardeni J, Meridor D, Kasher R, Nathan I, Parola AH. Humanin Derivatives Inhibit Necrotic Cell Death in Neurons. Mol Med 2015; 21:505-14. [PMID: 26062019 PMCID: PMC4607621 DOI: 10.2119/molmed.2015.00073] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/01/2015] [Indexed: 11/06/2022] Open
Abstract
Humanin and its derivatives are peptides known for their protective antiapoptotic effects against Alzheimer's disease. Herein, we identify a novel function of the humanin-derivative AGA(C8R)-HNG17 (namely, protection against cellular necrosis). Necrosis is one of the main modes of cell death, which was until recently considered an unmoderated process. However, recent findings suggest the opposite. We have found that AGA(C8R)-HNG17 confers protection against necrosis in the neuronal cell lines PC-12 and NSC-34, where necrosis is induced in a glucose-free medium by either chemohypoxia or by a shift from apoptosis to necrosis. Our studies in traumatic brain injury models in mice, where necrosis is the main mode of neuronal cell death, have shown that AGA(C8R)-HNG17 has a protective effect. This result is demonstrated by a decrease in a neuronal severity score and by a reduction in brain edema, as measured by magnetic resonance imaging (MRI). An insight into the peptide's antinecrotic mechanism was attained through measurements of cellular ATP levels in PC-12 cells under necrotic conditions, showing that the peptide mitigates a necrosis-associated decrease in ATP levels. Further, we demonstrate the peptide's direct enhancement of the activity of ATP synthase activity, isolated from rat-liver mitochondria, suggesting that AGA(C8R)-HNG17 targets the mitochondria and regulates cellular ATP levels. Thus, AGA(C8R)-HNG17 has potential use for the development of drug therapies for necrosis-related diseases, for example, traumatic brain injury, stroke, myocardial infarction, and other conditions for which no efficient drug-based treatment is currently available. Finally, this study provides new insight into the mechanisms underlying the antinecrotic mode of action of AGA(C8R)-HNG17.
Collapse
Affiliation(s)
- Aviv Cohen
- Department of Chemistry, The Faculty of Natural Sciences, Ben-Gurion University of the Negev, Be’er-Sheva, Israel
| | - Jenny Lerner-Yardeni
- Department of Chemistry, The Faculty of Natural Sciences, Ben-Gurion University of the Negev, Be’er-Sheva, Israel
| | - David Meridor
- Department of Chemistry, The Faculty of Natural Sciences, Ben-Gurion University of the Negev, Be’er-Sheva, Israel
| | - Roni Kasher
- Department of Desalination and Water Treatment, Zuckerberg Institute for Water Research, The Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Sede Boqer Campus, Midreshet Sede Boqer, Israel
| | - Ilana Nathan
- Department of Clinical Biochemistry and Pharmacology, The Faculty of Health Sciences, Ben-Gurion University of the Negev, Be’er-Sheva, Israel
- Institute of Hematology, Soroka University Medical Center, Be’er-Sheva, Israel
| | - Abraham H Parola
- Department of Chemistry, The Faculty of Natural Sciences, Ben-Gurion University of the Negev, Be’er-Sheva, Israel
| |
Collapse
|
39
|
Kumphune S, Surinkaew S, Chattipakorn SC, Chattipakorn N. Inhibition of p38 MAPK activation protects cardiac mitochondria from ischemia/reperfusion injury. PHARMACEUTICAL BIOLOGY 2015; 53:1831-1841. [PMID: 25880145 DOI: 10.3109/13880209.2015.1014569] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Cardiac cell death and fatal arrhythmias during myocardial ischemia/reperfusion (I/R) can be reduced by p38 MAPK inhibition. However, the effects of p38 MAPK inhibition on cardiac mitochondria have not been investigated. OBJECTIVE We tested the hypothesis that p38 MAPK inhibition at different times during I/R protects cardiac mitochondrial functions. MATERIALS AND METHODS Adult Wistar rats were subjected to 30 min of left anterior descending coronary artery (LAD) occlusion, followed by 120 min of reperfusion. A 2 mg/kg bolus infusion of p38 MAPK inhibitor, SB203580, was given before or during ischemia, or at reperfusion. Mitochondrial function and ultrastructure were assessed and Western blots were performed. RESULTS Administration of SB203580 at any time point of I/R significantly attenuated the mitochondrial ultrastructure change, mitochondrial swelling, by increasing the absorbance at 540 nm (I/R control 0.42 ± 0.03; pretreatment 0.58 ± 0.04; during ischemia 0.49 ± 0.02; at reperfusion 0.51 ± 0.02, p < 0.05), similar to reactive oxygen species (ROS) generation (I/R control 1300 ± 48; pretreatment 1150 ± 30; during ischemia 1000 ± 50; at reperfusion 1050 ± 55, p < 0.05). Only SB203580 given before or during ischemia attenuated mitochondrial membrane depolarization (I/R control 0.78 ± 0.04; pretreatment 1.02 ± 0.03; during ischemia 1.05 ± 0.12, p < 0.05). In addition, pre-treatment of SB203580 significantly reduced the phosphorylation of p53, CREB, Bax, cytochrome c, and cleaved caspase 3. DISCUSSION AND CONCLUSION The results from this study showed for the first time that p38 MAPK inhibition protects mitochondria from I/R injury.
Collapse
Affiliation(s)
- Sarawut Kumphune
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University , Chiang Mai , Thailand
| | | | | | | |
Collapse
|
40
|
Nozaki Y, Kinoshita K, Hino S, Yano T, Niki K, Hirooka Y, Kishimoto K, Funauchi M, Matsumura I. Signaling Rho-kinase mediates inflammation and apoptosis in T cells and renal tubules in cisplatin nephrotoxicity. Am J Physiol Renal Physiol 2015; 308:F899-909. [DOI: 10.1152/ajprenal.00362.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 01/20/2015] [Indexed: 11/22/2022] Open
Abstract
Nephrotoxicity is a frequent complication of cisplatin-induced chemotherapy, in which T cells are known to promote acute kidney injury (AKI). Apoptosis and necrosis of tubules and inflammatory events also contribute to the nephrotoxicity. A delineation of the mechanisms that underlie the inappropriate renal and tubular inflammation can thus provide important insights into potential therapies for cisplatin-induced AKI. Rho-kinases are known to act as molecular switches controlling several critical cellular functions, including cell migration, cytokine production, and apoptosis. Here, we show that the Rho-kinase inhibitor fasudil attenuated cisplatin nephrotoxicity, resulting in less histological damage, improved renal function, and the infiltration of fewer leukocytes into the kidney. Renal nuclear factor-κB activation and apoptosis were reduced, and the expressions of proinflammatory renal cytokine and chemokine mRNA were decreased. Urinary and renal kidney injury molecule-1 (Kim-1) expression was also reduced, a finding that is consistent with diminished kidney injury. In the current study, we also showed that fasudil could be protective of the impaired tubules. In vitro, fasudil reduced the apoptosis (annexin-V+PI cells) and cytokine production (tumor necrosis factor+ cells) in T cells and the apoptosis (annexin-V+PI cells) and tubular damage (Kim-1+ cells) in proximal tubular cells by flow cytometric analysis. As Rho-kinase plays an important role in promoting cisplatin nephrotoxicity, inhibiting Rho-kinase may be a therapeutic strategy for preventing cisplatin-induced AKI.
Collapse
Affiliation(s)
- Yuji Nozaki
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Koji Kinoshita
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Shoichi Hino
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Tomohiro Yano
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Kaoru Niki
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Yasuaki Hirooka
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Kazuya Kishimoto
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Masanori Funauchi
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kindai University School of Medicine, Osaka, Japan
| |
Collapse
|
41
|
Transient receptor potential melastatin-4 is involved in hypoxia-reoxygenation injury in the cardiomyocytes. PLoS One 2015; 10:e0121703. [PMID: 25836769 PMCID: PMC4383534 DOI: 10.1371/journal.pone.0121703] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 02/18/2015] [Indexed: 12/22/2022] Open
Abstract
Ischemic heart disease still remains the most common cause of cardiac death. During ischemia-reperfusion (I/R), reactive oxygen species (ROS) are produced in excess in cardiac tissue, where they induce cell death. Our previous study showed that 9-phenanthrol (9-Phe), a specific inhibitor of the TRPM4 channel, preserves cardiac contractile function and protects the heart from I/R injury-related infarction in the excised rat heart. Accordingly, we hypothesized that TRPM4 channels are involved in the 9-Phe-mediated cardioprotection against ROS-induced injury. In rats, intravenous 9-Phe mitigated the development of myocardial infarction caused by the occlusion of the left anterior descending artery. Immunohistochemical analysis indicated that TRPM4 proteins are expressed in ventricular myocytes susceptible to I/R injury. Hydrogen peroxide (H2O2) is among the main ROS overproduced during I/R. In the cardiomyocyte cell line H9c2, pretreatment with 9-Phe prevented cell death induced by conditions mimicking I/R, namely 200 μM H2O2 and hypoxia-reoxygenation. Gene silencing of TRPM4 preserved the viability of H9c2 cardiomyocytes exposed to 200 μM H2O2. These results suggest that the cardioprotective effects of 9-Phe are mediated through the inhibition of the TRPM4 channels.
Collapse
|
42
|
Abstract
Ischemia reperfusion injury occurs in the kidney when blood supply is interrupted in clinical settings such as kidney transplantation or nephron sparing surgery for renal tumors. These lesions lead to acute kidney injury (AKI) a detrimental situation associated with impaired short-term allograft function (delayed graft function or primary non function) but also long-term transplant survival through the onset of chronic allograft nephropathy. The present review details the cellular and molecular consequences of ischemia reperfusion in a native kidney as well as in a kidney graft after cold ischemia time, giving a comprehensive description of biological pathways involved during the phase of ischemia and during the reperfusion period where the rapid return to normoxia leads to a large burst of reactive oxygen species along with a dramatic reduction in antioxidant defenses. This work also focuses on the distinct susceptibilities of kidney cells to ischemia (endothelial vs epithelial) and the outcome of acute kidney injury.
Collapse
|
43
|
Biobanking: The Future of Cell Preservation Strategies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 864:37-53. [PMID: 26420612 DOI: 10.1007/978-3-319-20579-3_4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With established techniques cryopreservation is often viewed as an "old school" discipline yet modern cryopreservation is undergoing another scientific and technology development growth phase. In this regard, today's cryopreservation processes and cryopreserved products are found at the forefront of research in the areas of discovery science, stem cell research, diagnostic development and personalized medicine. As the utilization of cryopreserved cells continues to increase, the demands placed on the biobanking industry are increasing and evolving at an accelerated rate. No longer are samples providing for high immediate post-thaw viability adequate. Researchers are now requiring samples where not only is there high cell recovery but that the product recovered is physiologically and biochemically identical to its pre-freeze state at the genominic, proteomic, structural, functional and reproductive levels. Given this, biobanks are now facing the challenge of adapting strategies and protocols to address these needs moving forward. Recent studies have shown that the control and direction of the molecular response of cells to cryopreservation significantly impacts final outcome. This chapter provides an overview of the molecular stress responses of cells to cryopreservation, the impact of the apoptotic and necrotic cell death continuum and how studies focused on the targeted modulation of common and/or cell specific responses to freezing temperatures provide a path to improving sample quality and utility. This line of investigation has provided a new direction and molecular-based foundation guiding new research, technology development and procedures. As the use of and the knowledge base surrounding cryopreservation continues to expand, this path will continue to provide for improvements in overall efficacy and outcome.
Collapse
|
44
|
Bissoyi A, Nayak B, Pramanik K, Sarangi SK. Targeting cryopreservation-induced cell death: a review. Biopreserv Biobank 2014; 12:23-34. [PMID: 24620767 DOI: 10.1089/bio.2013.0032] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite marked developments in the field of cryopreservation of cells and tissues for research and therapeutic applications, post-thaw cell death remains a significant drawback faced by cryobiologists. Post cryopreservation apoptosis and necrosis are normally observed within 6 to 24 h after post-thaw culture. As a result, massive loss of cell viability and cellular function occur due to cryopreservation. However, in this new generation of cryopreservation science, scientists in this field are focusing on incorporation of apoptosis and necrosis inhibitors (zVAD-fmk, p38 MAPK inhibitor, ROCK inhibitor, etc.) to cryopreservation and post-thaw culture media. These inhibitors target and inhibit various proteins such as caspases, proteases, and kinases, involved in the cell death cascade, resulting in reduced intensity of apoptosis and necrosis in the cryopreserved cells and tissues, increased cell viability, and maintenance of cellular function; thus improved overall cryopreservation efficiency is achieved. The present article provides an overview of various cell death pathways, molecules mediating cryopreservation-induced apoptosis and the potential of certain molecules in targeting cryopreservation-induced delayed-onset cell death.
Collapse
Affiliation(s)
- A Bissoyi
- 1 Department of Biotechnology and Medical Engineering, National Institute of Technology , Rourkela, India
| | | | | | | |
Collapse
|
45
|
Ma Z, Wei Q, Dong G, Huo Y, Dong Z. DNA damage response in renal ischemia-reperfusion and ATP-depletion injury of renal tubular cells. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1088-96. [PMID: 24726884 PMCID: PMC4038345 DOI: 10.1016/j.bbadis.2014.04.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/25/2014] [Accepted: 04/02/2014] [Indexed: 12/12/2022]
Abstract
Renal ischemia-reperfusion leads to acute kidney injury (AKI) that is characterized pathologically by tubular damage and cell death, followed by tubular repair, atrophy and interstitial fibrosis. Recent work suggested the possible presence of DNA damage response (DDR) in AKI. However, the evidence is sketchy and the role and regulation of DDR in ischemic AKI remain elusive. In this study, we demonstrated the induction of phosphorylation of ATM, H2AX, Chk2 and p53 during renal ischemia-reperfusion in mice, suggesting DDR in kidney tissues. DDR was also induced in vitro during the recovery or "reperfusion" of renal proximal tubular cells (RPTCs) after ATP depletion. DDR in RPTCs was abrogated by supplying glucose to maintain ATP via glycolysis, indicating that the DDR depends on ATP depletion. The DDR was also suppressed by the general caspase inhibitor z-VAD and the overexpression of Bcl-2, supporting a role of apoptosis-associated DNA damage in the DDR. N-acetylcysteine (NAC), an antioxidant, suppressed the phosphorylation of ATM and p53 and, to a less extent, Chk2, but NAC increased the phosphorylation and nuclear foci formation of H2AX. Interestingly, NAC increased apoptosis, which may account for the observed H2AX activation. Ku55933, an ATM inhibitor, blocked ATM phosphorylation and ameliorated the phosphorylation of Chk2 and p53, but it increased H2AX phosphorylation and nuclear foci formation. Ku55933 also increased apoptosis in RPTCs following ATP depletion. The results suggest that DDR occurs during renal ischemia-reperfusion in vivo and ATP-depletion injury in vitro. The DDR is partially induced by apoptosis and oxidative stress-related DNA damage. ATM, as a sensor in the DDR, may play a cytoprotective role against tubular cell injury and death.
Collapse
Affiliation(s)
- Zhengwei Ma
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Reagents University and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Reagents University and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Reagents University and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Georgia Reagents University and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Reagents University and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA; Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
46
|
Salutary effect of calcium channel blockade following hypoxic and septic insult. J Trauma Acute Care Surg 2014; 77:40-6; discussion 45-6. [PMID: 24977753 DOI: 10.1097/ta.0000000000000260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Intestinal ischemia and reperfusion is a major problem associated with a high morbidity and mortality following trauma and hemorrhagic shock. Apoptosis is the major mode of cell death following reperfusion. The cytoskeleton damage precedes the apoptotic final microscopic features. Calcium plays a central role in apoptosis. Therefore, we studied whether verapamil could preserve the function of the cytoskeleton in an in vitro intestinal model following hypoxia-reoxygenation (H/R). Our goal was to assess mainly the cytoskeleton functions, which includes IgA transport and the cell monolayer barrier integrity. METHODS Confluent HT29 intestinal monolayers grown in a two-chamber cell culture system were held under hypoxic (5% CO2) conditions for 90 minutes followed by normoxia (21% O2) (H/R). Cell subsets were exposed to lipopolysaccharide (10 μg/mL) before H/R. Verapamil (8 μM) was added to HT29 cell subsets after H/R treatment. Dimeric IgA was added to the basal compartment, and apical media were sampled at intervals to quantitate IgA transcytosis using enzyme-linked immunosorbent assay. HT29 cells held under normoxic conditions served as controls. HT29 permeability to FD4 was assessed at the end of each experiment. In a separate experiment, HT29 cells were stained for F actin using rhodamine-labeled phalloidin. RESULTS Intestinal monolayer permeability was increased following treatment with H/R and/or lipopolysaccharide. Verapamil treatment prevented increased permeability in HT29 cells and led to an increase in IgA transport. Disruption of actin microfilaments was demonstrated following H/R insult but was abrogated by the addition of verapamil following H/R insult. CONCLUSION Reperfusion can lead to both physical and immune derangement of epithelial cell barrier function. Verapamil may be important in preserving gut barrier function. Additional studies including in vivo confirmation in animal shock models are needed to validate these findings.
Collapse
|
47
|
Abstract
BACKGROUND Generation of reactive oxygen species (ROS) is an important mechanism of ischemia-reperfusion injury. Abrupt reoxygenation compared with slow reoxygenation has been known to increase ROS generation. Thus, slow and stepwise reperfusion can reduce ROS generation and subsequent ischemia-reperfusion injury. This study investigated the effect of slow reperfusion by blood pressure-targeted stepwise resuscitation (PSR) in hemorrhagic shock. METHODS Pressure-controlled hemorrhagic shock was induced in male Sprague-Dawley rats for 1 hour. Rats were then allocated to one of three groups (no-resuscitation group, n = 14; PSR group, n = 15; rapid normalization of blood pressure (RR) group, n = 15). Survival time and hemodynamic changes were recorded and compared. Blood samples and liver tissue were harvested after 6 hours of resuscitation in surviving rats. RESULTS All of the rats in the no-resuscitation group were expired before the end of the 6-hour observation period. Survival times were significantly longer in the PSR group than in the RR group (survival rates, 11 of 15 vs. 5 of 15, log rank p = 0.032). Plasma amino alanine transferase, histologic liver injury, and ROS generation in the liver tissue were significantly lower in the PSR group than in the RR group (all findings significant, p < 0.05). In addition, PSR significantly decreased plasma nitric oxide, liver interleukin 1β, and liver interleukin 6 compared with rapid resuscitation in addition to augmenting Akt survival pathways (all p < 0.05). CONCLUSION Slow reperfusion by PSR decreased mortality, ROS generation, and liver injury in rats undergoing hemorrhagic shock. Stepwise resuscitation also decreased inflammatory cytokine production and augmented Akt survival pathways.
Collapse
|
48
|
Selective 14-3-3γ induction quenches p-β-catenin Ser37/Bax-enhanced cell death in cerebral cortical neurons during ischemia. Cell Death Dis 2014; 5:e1184. [PMID: 24743739 PMCID: PMC4001306 DOI: 10.1038/cddis.2014.152] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 02/20/2014] [Accepted: 03/07/2014] [Indexed: 01/05/2023]
Abstract
Ischemia-induced cell death is a major cause of disability or death after stroke. Identifying the key intrinsic protective mechanisms induced by ischemia is critical for the development of effective stroke treatment. Here, we reported that 14-3-3γ was a selective ischemia-inducible survival factor in cerebral cortical neurons reducing cell death by downregulating Bax depend direct 14-3-3γ/p-β-catenin Ser37 interactions in the nucleus. 14-3-3γ, but not other 14-3-3 isoforms, was upregulated in primary cerebral cortical neurons upon oxygen–glucose deprivation (OGD) as measured by quantitative PCR, western blot and fluorescent immunostaining. The selective induction of 14-3-3γ in cortical neurons by OGD was verified by the in vivo ischemic stroke model. Knocking down 14-3-3γ alone or inhibiting 14-3-3/client interactions was sufficient to induce cell death in normal cultured neurons and exacerbate OGD-induced neuronal death. Ectopic overexpression of 14-3-3γ significantly reduced OGD-induced cell death in cultured neurons. Co-immunoprecipitation and fluorescence resonance energy transfer demonstrated that endogenous 14-3-3γ bound directly to more p-β-catenin Ser37 but not p-Bad, p-Ask-1, p-p53 and Bax. During OGD, p-β-catenin Ser37 but not p-β-catenin Ser45 was increased prominently, which correlated with Bax elevation in cortical neurons. OGD promoted the entry of 14-3-3γ into the nuclei, in correlation with the increase of nuclear p-β-catenin Ser37 in neurons. Overexpression of 14-3-3γ significantly reduced Bax expression, whereas knockdown of 14-3-3γ increased Bax in cortical neurons. Abolishing β-catenin phosphorylation at Ser37 (S37A) significantly reduced Bax and cell death in neurons upon OGD. Finally, 14-3-3γ overexpression completely suppressed β-catenin-enhanced Bax and cell death in neurons upon OGD. Based on these data, we propose that the 14-3-3γ/p-β-catenin Ser37/Bax axis determines cell survival or death of neurons during ischemia, providing novel therapeutic targets for ischemic stroke as well as other related neurological diseases.
Collapse
|
49
|
da Cunha MJ, da Cunha AA, Scherer EBS, Machado FR, Loureiro SO, Jaenisch RB, Guma F, Lago PD, Wyse ATS. Experimental lung injury promotes alterations in energy metabolism and respiratory mechanics in the lungs of rats: prevention by exercise. Mol Cell Biochem 2013; 389:229-38. [PMID: 24378995 DOI: 10.1007/s11010-013-1944-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 12/18/2013] [Indexed: 12/11/2022]
Abstract
In the present study we investigated the effects of lung injury on energy metabolism (succinate dehydrogenase, complex II, cytochrome c oxidase, and ATP levels), respiratory mechanics (dynamic and static compliance, elastance and respiratory system resistance) in the lungs of rats, as well as on phospholipids in bronchoalveolar lavage fluid. The protective effect of physical exercise on the alterations caused by lung injury, including lung edema was also evaluated. Wistar rats were submitted to 2 months of physical exercise. After this period the lung injury was induced by intratracheal instillation of lipopolysaccharide. Adult Wistar rats were submitted to 2 months of physical exercise and after this period the lung injury was induced by intratracheal instillation of lipopolysaccharide in dose 100 μg/100 g body weight. The sham group received isotonic saline instillation. Twelve hours after the injury was performed the respiratory mechanical and after the rats were decapitated and samples were collected. The rats subjected to lung injury presented a decrease in activities of the enzymes of the electron transport chain and ATP levels in lung, as well as the formation of pulmonary edema. A decreased lung dynamic and static compliance, as well as an increase in respiratory system resistance, and a decrease in phospholipids content were observed. Physical exercise was able to totally prevent the decrease in succinate dehydrogenase and complex II activities and the formation of pulmonary edema. It also partially prevented the increase in respiratory system resistance, but did not prevent the decrease in dynamic and static compliance, as well as in phospholipids content. These findings suggest that the mitochondrial dysfunction may be one of the important contributors to lung damage and that physical exercise may be beneficial in this pathology, although it did not prevent all changes present in lung injury.
Collapse
Affiliation(s)
- Maira J da Cunha
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, ICBS, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ojeda-Cervantes M, Barrera-Chimal J, Alberú J, Pérez-Villalva R, Morales-Buenrostro LE, Bobadilla NA. Mineralocorticoid receptor blockade reduced oxidative stress in renal transplant recipients: a double-blind, randomized pilot study. Am J Nephrol 2013; 37:481-90. [PMID: 23635604 DOI: 10.1159/000350539] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 03/06/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Previous experimental studies from our laboratory have demonstrated that aldosterone plays a central role in renal ischemic processes. This study was designed to evaluate the effect of mineralocorticoid receptor blockade in renal transplant recipients from living donors. METHODS 20 adult kidney transplant recipients from living donors were included in a double-blind, randomized, placebo-controlled clinical pilot study that compared spironolactone and placebo. Placebo or spironolactone (25 mg) was administered 1 day before and 3 days posttransplantation. Renal function and urinary kidney injury molecule-1, interleukin-18, and heat shock protein 72 as well as urinary hydrogen peroxide (H2O2) levels were quantified. RESULTS No significant differences were seen between the groups studied regarding age, gender, indication for kidney transplantation, residual renal function, renal replacement therapy, or warm and cold ischemia periods. In contrast, spironolactone administration significantly reduced the oxidative stress assessed by the urinary H2O2 excretion, in spite of no differences in renal function or reduction in tubular injury biomarkers. CONCLUSIONS The findings of this exploratory study strongly suggest that aldosterone promotes oxidative stress and that the administration of spironolactone reduces the production of urinary H2O2 as a result of lesser formation of surrogate reactive oxygen species secondary to the ischemia-reperfusion phenomenon.
Collapse
Affiliation(s)
- Marcos Ojeda-Cervantes
- Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | | | | | | | | |
Collapse
|