301
|
Chimeh U, Zimmerman MA, Gilyazova N, Li PA. B355252, A Novel Small Molecule, Confers Neuroprotection Against Cobalt Chloride Toxicity In Mouse Hippocampal Cells Through Altering Mitochondrial Dynamics And Limiting Autophagy Induction. Int J Med Sci 2018; 15:1384-1396. [PMID: 30275767 PMCID: PMC6158673 DOI: 10.7150/ijms.24702] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/12/2018] [Indexed: 12/31/2022] Open
Abstract
Cerebral hypoxia as often occurs in cases of stroke, hemorrhage, or other traumatic brain injuries, is one of the leading causes of death worldwide and a main driver of disabilities in the elderly. Using a chemical mimetic of hypoxia, cobalt chloride (CoCl2), we tested the ability of a novel small molecule, 4-chloro-N-(naphthalen-1-ylmethyl)-5-(3-(piperazin-1-yl)phenoxy)thiophene-2-sulfonamide (B355252), to alleviate CoCl2-induced damage in mouse hippocampal HT22 cells. A dose-dependent decrease in cell viability was observed during CoCl2 treatment along with increases in mitochondrial membrane potential and generation of reactive oxygen species (ROS). B355252 conferred protection against these changes. We further found that mitochondrial dynamics, the balance between mitochondrial fusion and fission, were perturbed by CoCl2 treatment. Mitochondrial fusion, which was assessed by measuring the expression of proteins optic atrophy protein 1 (OPA1) and mitofusin 2 (Mfn2), declined due to CoCl2 exposure, but B355252 addition was able to elevate Mfn2 expression while OPA1 expression was unchanged. Mitochondrial fission, measured by phosphorylated dynamin-related protein 1 (p-DRP1) and fission protein 1 (FIS1) expression, also decreased following CoCl2 exposure, and was stabilized by B355252 addition. Finally, autophagy was assessed by measuring the conversion of cytosolic microtubule-associated protein 1A/1B-light chain three-I (LC3-I) to autophagosome-bound microtubule-associated protein 1A/1B-light chain three-II (LC3-II) and was found to be increased by CoCl2. B355252 addition significantly reduced autophagy induction. Taken together, our results indicate B355252 has therapeutic potential to reduce the damaging effects caused by CoCl2 and should be further evaluated for applications in cerebral ischemia therapy.
Collapse
Affiliation(s)
| | | | | | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC USA
| |
Collapse
|
302
|
Rana AK, Singh D. Targeting glycogen synthase kinase-3 for oxidative stress and neuroinflammation: Opportunities, challenges and future directions for cerebral stroke management. Neuropharmacology 2018; 139:124-136. [DOI: 10.1016/j.neuropharm.2018.07.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/02/2018] [Accepted: 07/05/2018] [Indexed: 12/15/2022]
|
303
|
Shah FA, Park DJ, Koh PO. Identification of Proteins Differentially Expressed by Quercetin Treatment in a Middle Cerebral Artery Occlusion Model: A Proteomics Approach. Neurochem Res 2018; 43:1608-1623. [DOI: 10.1007/s11064-018-2576-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 06/06/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023]
|
304
|
Sarmah D, Kaur H, Saraf J, Vats K, Pravalika K, Wanve M, Kalia K, Borah A, Kumar A, Wang X, Yavagal DR, Dave KR, Bhattacharya P. Mitochondrial Dysfunction in Stroke: Implications of Stem Cell Therapy. Transl Stroke Res 2018; 10:10.1007/s12975-018-0642-y. [PMID: 29926383 DOI: 10.1007/s12975-018-0642-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 05/21/2018] [Accepted: 06/12/2018] [Indexed: 01/06/2023]
Abstract
Stroke is a debilitating condition which is also the second leading cause of death and disability worldwide. Despite the benefits and promises shown by numerous neuroprotective agents in animal stroke models, their clinical translation has not been a complete success. Hence, search for treatment options have directed researchers towards utilising stem cells. Mitochondria has a major involvement in the pathophysiology of stroke and a number of other conditions. Stem cells have shown the ability to transfer mitochondria to the damaged cells and to help revive cell energetics in the recipient cell. The present review discusses how stem cells could be employed to protect neurons and mitochondria in stroke and also the various mechanisms involved in neuroprotection.
Collapse
Affiliation(s)
- Deepaneeta Sarmah
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Harpreet Kaur
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Jackson Saraf
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kanchan Vats
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kanta Pravalika
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Madhuri Wanve
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kiran Kalia
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Akhilesh Kumar
- Department of Botany, Banaras Hindu University, Varanasi, India
| | - Xin Wang
- Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Dileep R Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Pallab Bhattacharya
- Department or Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
305
|
Ryu EJ, Kim DW, Shin MJ, Jo HS, Park JH, Cho SB, Lee CH, Yeo HJ, Yeo EJ, Choi YJ, Kim DS, Cho SW, Cho YJ, Sohn EJ, Son O, Lee KW, Han KH, Park J, Eum WS, Choi SY. PEP‑1‑glutaredoxin 1 protects against hippocampal neuronal cell damage from oxidative stress via regulation of MAPK and apoptotic signaling pathways. Mol Med Rep 2018; 18:2216-2228. [PMID: 29916538 DOI: 10.3892/mmr.2018.9176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 01/11/2018] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress is known to be a primary risk factor for neuronal diseases. Glutaredoxin (GLRX)‑1, a redox‑regulator of the thioredoxin superfamily, is known to exhibit an important role in cell survival via various cellular functions. However, the precise roles of GLRX1 in brain ischemia are still not fully understood. The present study investigated whether transduced PEP‑1‑GLRX1 protein has protective effects against oxidative stress in cells and in an animal model. Transduced PEP‑1‑GLRX1 protein increased HT‑22 cell viability under oxidative stress and this fusion protein significantly reduced intracellular reactive oxygen species and levels of DNA damage. In addition, PEP‑1‑GLRX1 protein regulated RAC‑a serine/threonine‑protein kinase and mitogen‑activated protein kinase signaling, in addition to apoptotic signaling including B cell lymphoma (Bcl)‑2, Bcl‑2 associated X, apoptosis regulator, pro‑caspase‑9 and p53 expression levels. In an ischemic animal model, it was verified that PEP‑1‑GLRX1 transduced into the Cornu Ammonis 1 region of the animal brain, where it markedly protected against ischemic injury. These results indicate that PEP‑1‑GLRX1 attenuates neuronal cell death resulting from oxidative stress in vitro and in vivo. Therefore, PEP‑1‑GLRX1 may exhibit a beneficial role in the treatment of neuronal disorders, including ischemic injury.
Collapse
Affiliation(s)
- Eun Ji Ryu
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung‑Wonju National University, Gangneung, Gangwon 25457, Republic of South Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Hyo Sang Jo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Su Bin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Chi Hern Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan‑Si, South Chungcheong 31538, Republic of South Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Republic of South Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University Medical Center, Chuncheon, Gangwon 24253, Republic of South Korea
| | - Eun Jeong Sohn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Ora Son
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of South Korea
| |
Collapse
|
306
|
Shi X, Liu HY, Li SP, Xu HB. Keratinocyte growth factor protects endometrial cells from oxygen glucose deprivation/re-oxygenation via activating Nrf2 signaling. Biochem Biophys Res Commun 2018; 501:178-185. [DOI: 10.1016/j.bbrc.2018.04.208] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022]
|
307
|
Squitti R, Siotto M, Assenza G, Giannantoni NM, Rongioletti M, Zappasodi F, Tecchio F. Prognostic Value of Serum Copper for Post-Stroke Clinical Recovery: A Pilot Study. Front Neurol 2018; 9:333. [PMID: 29899723 PMCID: PMC5988843 DOI: 10.3389/fneur.2018.00333] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022] Open
Abstract
The clinical course after ischemic stroke can vary considerably despite similar lesions and clinical status at the onset of symptoms, suggesting that individual factors modulate clinical recovery. Here, we sought to test the working hypothesis that elevated copper values provide prognostic information, and specifically predict worse clinical recovery. We further sought to support previous findings regarding metal metabolism in acute stroke. We assessed total antioxidant status, oxidative stress factors (peroxides) and metal metabolism markers (iron, copper, ceruloplasmin concentration and activity, ferritin, and transferrin) in the acute phase (2–10 days from symptom onset) in 30 patients affected by unilateral middle cerebral artery (MCA) stroke. A longitudinal assessment of clinical deficit was performed in the acute and stabilized phases (typically 6 months post-stroke) using the National Institutes of Health Stroke Scale (NIHSS). In identifying recovery-related factors, we considered effective recovery (ER), calculated as the ratio between actual NIHSS recovery and the total potential recovery. This allows an estimation of the actual recovery adjusted for the patient’s initial condition. In the acute phase, clinical severity was correlated with increased peroxide concentrations, and lower iron levels. Less successful clinical recovery was correlated with increased acute copper levels, which entered a multiple regression model that explained 24% of ER variance. These pilot data suggest that, in the acute phase of an ischemic stroke, copper may provide useful information about clinical recovery.
Collapse
Affiliation(s)
- Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Giovanni Assenza
- Clinical Neurology, Campus Biomedico University of Rome, Rome, Italy
| | - Nadia M Giannantoni
- Neurocenter of Southern Switzerland, Civic Hospital, Lugano, Switzerland.,Laboratory of Electrophysiology for Translational neuroScience (LET'S), ISTC-CNR, Rome, Italy
| | - Mauro Rongioletti
- Department of Biology Medicine, Research and Development Division, Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy
| | - Filippo Zappasodi
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Institute for Advanced Biomedical Technologies, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Franca Tecchio
- Laboratory of Electrophysiology for Translational neuroScience (LET'S), ISTC-CNR, Rome, Italy.,Institute of Neurology, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
308
|
Synaptic transmission and excitability during hypoxia with inflammation and reoxygenation in hippocampal CA1 neurons. Neuropharmacology 2018; 138:20-31. [PMID: 29775678 DOI: 10.1016/j.neuropharm.2018.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/19/2018] [Accepted: 05/08/2018] [Indexed: 12/30/2022]
Abstract
Although a number of experimental and clinical studies have shown that hypoxia typically accompanies acute inflammatory responses, the combinatorial effect of the two insults on basic neural function has not been thoroughly investigated. Previous studies have predominantly suggested that hypoxia reduces network activity; however, several studies suggest the opposite effect. Of note, inflammation is known to increase neural activity. In the current study, we examined the effects of limited oxygen in combination with an inflammatory stimulus, as well as the effects of reoxygenation, on synaptic transmission and excitability. We observed a significant reduction of both synaptic transmission and excitability when hypoxia and inflammation occurred in combination, whereas reoxygenation caused hyperexcitability of neurons. Further, we found that the observed reduction in synaptic transmission was due to compromised presynaptic release efficiency based on an adenosine-receptor-dependent increase in synaptic facilitation. Excitability changes in both directions were attributable to dynamic regulation of the hyperpolarization-activated cation current (Ih) and to changes in the input resistance and the voltage difference between resting membrane potential and action potential threshold. We found that zatebradine, an Ih current inhibitor, reduced the fluctuation in excitability, suggesting that it may have potential as a drug to ameliorate reperfusion brain injury.
Collapse
|
309
|
Metabolic influence of walnut phenolic extract on mitochondria in a colon cancer stem cell model. Eur J Nutr 2018; 58:1635-1645. [PMID: 29740695 DOI: 10.1007/s00394-018-1708-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 05/01/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Walnut phenolic extract (WPE) reduces proliferation and enhances differentiation of colon cancer stem cells (CSCs). The present study investigated the metabolic influence of WPE on the mitochondrial function of colon CSCs to determine its underlying mechanism. METHODS CD133+CD44+ HCT116 colon cancer cells were selected by fluorescence-activated cell sorting and were treated with or without 40 µg/mL WPE. RNA-sequencing (RNA-Seq) was performed to identify differentially expressed genes (DEGs), which were further validated with RT-PCR. WPE-induced alterations in mitochondrial function were investigated through a mitochondrial stress test by determining cellular oxygen consumption rate (OCR), an indicator of mitochondrial respiration, and extracellular acidification rate (ECAR), an indicator of glycolysis, which were further confirmed by glucose uptake and lactate production tests. RESULTS RNA-Seq analysis identified two major functional clusters: metabolic and mitochondrial clusters. WPE treatment shifted the metabolic profile of cells towards the glycolysis pathway (ΔECAR = 36.98 mpH/min/ptn, p = 0.02) and oxidative pathway (ΔOCR = 29.18 pmol/min/ptn, p = 0.00001). Serial mitochondrial stimulations using respiration modulators, oligomycin, carbonyl cyanide-4 (trifluoromethoxy) phenylhydrazone, and rotenone/antimycin A, found an increased potential of mitochondrial respiration (ΔOCR = 111.5 pmol/min/ptn, p = 0.0006). WPE treatment also increased glucose uptake (Δ = 0.39 pmol/µL, p = 0.002) and lactate production (Δ = 0.08 nmol/µL, p = 0.005). CONCLUSIONS WPE treatment shifts the mitochondrial metabolism of colon CSC towards more aerobic glycolysis, which might be associated with the alterations in the characteristics of colon CSC.
Collapse
|
310
|
Waddington DEJ, Sarracanie M, Salameh N, Herisson F, Ayata C, Rosen MS. An Overhauser-enhanced-MRI platform for dynamic free radical imaging in vivo. NMR IN BIOMEDICINE 2018; 31:e3896. [PMID: 29493032 DOI: 10.1002/nbm.3896] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 06/08/2023]
Abstract
Overhauser-enhanced MRI (OMRI) is an electron-proton double-resonance imaging technique of interest for its ability to non-invasively measure the concentration and distribution of free radicals. In vivo OMRI experiments are typically undertaken at ultra-low magnetic field (ULF), as both RF power absorption and penetration issues-a consequence of the high resonance frequencies of electron spins-are mitigated. However, working at ULF causes a drastic reduction in MRI sensitivity. Here, we report on the design, construction and performance of an OMRI platform optimized for high NMR sensitivity and low RF power absorbance, exploring challenges unique to probe design in the ULF regime. We use this platform to demonstrate dynamic imaging of TEMPOL in a rat model. The work presented here demonstrates improved speed and sensitivity of in vivo OMRI, extending the scope of OMRI to the study of dynamic processes such as metabolism.
Collapse
Affiliation(s)
- David E J Waddington
- A. A. Martinos Center for Biomedical Imaging, 149 Thirteenth St., Charlestown, MA 02129, USA
- Department of Physics, Harvard University, 17 Oxford St, Cambridge, MA 02138, USA
- ARC Centre of Excellence for Engineered Quantum Systems, School of Physics, The University of Sydney, Sydney, NSW 2006, Australia
| | - Mathieu Sarracanie
- A. A. Martinos Center for Biomedical Imaging, 149 Thirteenth St., Charlestown, MA 02129, USA
- Department of Physics, Harvard University, 17 Oxford St, Cambridge, MA 02138, USA
- Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA
| | - Najat Salameh
- A. A. Martinos Center for Biomedical Imaging, 149 Thirteenth St., Charlestown, MA 02129, USA
- Department of Physics, Harvard University, 17 Oxford St, Cambridge, MA 02138, USA
- Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA
| | - Fanny Herisson
- Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA 02129, USA
| | - Cenk Ayata
- Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA
- Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA 02129, USA
| | - Matthew S Rosen
- A. A. Martinos Center for Biomedical Imaging, 149 Thirteenth St., Charlestown, MA 02129, USA
- Department of Physics, Harvard University, 17 Oxford St, Cambridge, MA 02138, USA
- Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA
| |
Collapse
|
311
|
Hentia C, Rizzato A, Camporesi E, Yang Z, Muntean DM, Săndesc D, Bosco G. An overview of protective strategies against ischemia/reperfusion injury: The role of hyperbaric oxygen preconditioning. Brain Behav 2018; 8:e00959. [PMID: 29761012 PMCID: PMC5943756 DOI: 10.1002/brb3.959] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 02/12/2018] [Accepted: 02/18/2018] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Ischemia/reperfusion (I/R) injury, such as myocardial infarction, stroke, and peripheral vascular disease, has been recognized as the most frequent causes of devastating disorders and death currently. Protective effect of various preconditioning stimuli, including hyperbaric oxygen (HBO), has been proposed in the management of I/R. METHODS In this study, we searched and reviewed up-to-date published papers to explore the pathophysiology of I/R injury and to understand the mechanisms underlying the protective effect of HBO as conditioning strategy. RESULTS Animal study and clinic observation support the notion that HBO therapy and conditioning provide beneficial effect against the deleterious effects of postischemic reperfusion. Several explanations have been proposed. The first likely mechanism may be that HBO counteracts hypoxia and reduces I/R injury by improving oxygen delivery to an area with diminished blood flow. Secondly, by reducing hypoxia-ischemia, HBO reduces all the pathological events as a consequence of hypoxia, including tissue edema, increased affective area permeability, postischemia derangement of tissue metabolism, and inflammation. Thirdly, HBO may directly affect cell apoptosis, signal transduction, and gene expression in those that are sensitive to oxygen or hypoxia. HBO provides a reservoir of oxygen at cellular level not only carried by blood, but also by diffusion from the interstitial tissue where it reaches high concentration that may last for several hours, improves endothelial function and rheology, and decreases local inflammation and edema. CONCLUSION Evidence suggests the benefits of HBO when used as a preconditioning stimulus in the setting of I/R injury. Translating the beneficial effects of HBO into current practice requires, as for the "conditioning strategies", a thorough consideration of risk factors, comorbidities, and comedications that could interfere with HBO-related protection.
Collapse
Affiliation(s)
- Ciprian Hentia
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy.,Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Alex Rizzato
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy
| | | | - Zhongjin Yang
- The Institute for Human Performance SUNY Upstate Medical University Syracuse NY USA
| | - Danina M Muntean
- Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania.,Center for Translational Research and Systems Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Dorel Săndesc
- Faculty of Medicine "Victor Babeș" University of Medicine and Pharmacy Timișoara Romania
| | - Gerardo Bosco
- Master II level in Hyperbaric Medicine Department of Biomedical Sciences University of Padova Padova Italy
| |
Collapse
|
312
|
Yin B, Barrionuevo G, Weber SG. Mitochondrial GSH Systems in CA1 Pyramidal Cells and Astrocytes React Differently during Oxygen-Glucose Deprivation and Reperfusion. ACS Chem Neurosci 2018; 9:738-748. [PMID: 29172440 DOI: 10.1021/acschemneuro.7b00369] [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] [Indexed: 11/29/2022] Open
Abstract
Pyramidal cells and astrocytes have differential susceptibility to oxygen-glucose deprivation and reperfusion (OGD-RP). It is known that excessive reactive oxygen species (ROS) in mitochondria initiates cell death, while glutathione (GSH) is one of the major defenses against ROS. Although it is known that astrocytes contain a higher concentration of GSH than neurons, and that astrocytes can provide neurons with GSH, we are unaware of a detailed and quantitative examination of the dynamic changes in the mitochondrial GSH system in the two cell types during OGD-RP. Here, we determined mitochondrial membrane potential and the degrees of oxidation of the mitochondrially targeted roGFP-based sensors for hydrogen peroxide (OxDP) and GSH (OxDG). We also developed a method to estimate the mitochondrial GSH (mGSH) concentration in single cells in the CA1 region of organotypic hippocampal slice cultures at several time-points during OGD-RP. We find that mitochondrial membrane potential drops in pyramidal cells during OGD while it is relatively stable in astrocytes. In both types of cell, the mitochondrial membrane potential decreases during RP. During OGD-RP, mitochondrial peroxide levels are the same. Astrocytic mGSH is more than four times higher than pyramidal cell mGSH (3.2 vs 0.7 mM). Astrocytic mGSH is drained from mitochondria during OGD, whereas in pyramidal cells it remains fairly constant. OxDGSH prior to and during OGD is lower (less oxidized) in pyramidal cells than in astrocytes, but the two nearly converge during RP. The larger changes of redox status in the GSH system in pyramidal cells than astrocytes is an upstream sign of the higher mortality of the pyramidal cells after facing an insult. The pattern of [mGSH] changes in the two cell types could be recognized as another mechanism by which astrocytes protect neurons from transient, extreme conditions.
Collapse
Affiliation(s)
- Bocheng Yin
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Germán Barrionuevo
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen G. Weber
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
313
|
Abutarboush R, Mullah SH, Saha BK, Haque A, Walker PB, Aligbe C, Pappas G, Tran Ho LTV, Arnaud FG, Auker CR, McCarron RM, Scultetus AH, Moon-Massat P. Brain oxygenation with a non-vasoactive perfluorocarbon emulsion in a rat model of traumatic brain injury. Microcirculation 2018; 25:e12441. [DOI: 10.1111/micc.12441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Rania Abutarboush
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Saad H. Mullah
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Biswajit K. Saha
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Ashraful Haque
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Peter B. Walker
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Chioma Aligbe
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | - Georgina Pappas
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | | | - Francoise G. Arnaud
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | - Charles R. Auker
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| | - Richard M. McCarron
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | - Anke H. Scultetus
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
- Department of Surgery; Uniformed Services University of the Health Sciences; Bethesda MD USA
| | - Paula Moon-Massat
- NeuroTrauma Department; Naval Medical Research Center; Silver Spring MD USA
| |
Collapse
|
314
|
Pak O, Sydykov A, Kosanovic D, Schermuly RT, Dietrich A, Schröder K, Brandes RP, Gudermann T, Sommer N, Weissmann N. Lung Ischaemia-Reperfusion Injury: The Role of Reactive Oxygen Species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:195-225. [PMID: 29047088 DOI: 10.1007/978-3-319-63245-2_12] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lung ischaemia-reperfusion injury (LIRI) occurs in many lung diseases and during surgical procedures such as lung transplantation. The re-establishment of blood flow and oxygen delivery into the previously ischaemic lung exacerbates the ischaemic injury and leads to increased microvascular permeability and pulmonary vascular resistance as well as to vigorous activation of the immune response. These events initiate the irreversible damage of the lung with subsequent oedema formation that can result in systemic hypoxaemia and multi-organ failure. Alterations in the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) have been suggested as crucial mediators of such responses during ischaemia-reperfusion in the lung. Among numerous potential sources of ROS/RNS within cells, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, xanthine oxidases, nitric oxide synthases and mitochondria have been investigated during LIRI. Against this background, we aim to review here the extensive literature about the ROS-mediated cellular signalling during LIRI, as well as the effectiveness of antioxidants as treatment option for LIRI.
Collapse
Affiliation(s)
- Oleg Pak
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Djuro Kosanovic
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Alexander Dietrich
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336, Munich, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Thomas Gudermann
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336, Munich, Germany
| | - Natascha Sommer
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany.
| |
Collapse
|
315
|
Nuñez A, Benavente I, Blanco D, Boix H, Cabañas F, Chaffanel M, Fernández-Colomer B, Fernández-Lorenzo JR, Loureiro B, Moral MT, Pavón A, Tofé I, Valverde E, Vento M. Oxidative stress in perinatal asphyxia and hypoxic-ischaemic encephalopathy. An Pediatr (Barc) 2018. [DOI: 10.1016/j.anpede.2017.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
316
|
Ginkgolide K attenuates neuronal injury after ischemic stroke by inhibiting mitochondrial fission and GSK-3β-dependent increases in mitochondrial membrane permeability. Oncotarget 2018; 8:44682-44693. [PMID: 28591721 PMCID: PMC5546510 DOI: 10.18632/oncotarget.17967] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/27/2017] [Indexed: 12/21/2022] Open
Abstract
Ginkgolide K (GK) belongs to the ginkgolide family of natural compounds found in Ginkgo biloba leaves, which have been used for centuries to treat cerebrovascular and cardiovascular diseases. We evaluated the protective effects of GK against neuronal apoptosis by assessing its ability to sustain mitochondrial integrity and function. Co-immunoprecipitation showed that Drp1 binding to GSK-3β was increased after an oxygen-glucose deprivation/reperfusion (OGD/R) insult in cultured neuroblastoma cells. This induced Drp1 and GSK-3β translocation to mitochondria and mitochondrial dysfunction, which was attenuated by GK. GK also reduced mitochondrial fission by increasing Drp1 phosphorylation at Ser637 and inhibiting mitochondrial Drp1 recruitment. In addition, GK exposure induced GSK-3β phosphorylation at Ser9 and enhanced the interaction between adenine nucleotide translocator (ANT) and p-GSK-3β. This interaction suppressed the interaction between ANT and cyclophilin D (CypD), which inhibited mitochondrial permeability transition pore (mPTP) opening. Similarly, suppression of mitochondrial fission by Mdivi-1 also inhibited GSK-3β-induced mPTP opening. Treating mice with GK prevented GSK-3β and Drp1 translocation to mitochondria and attenuated mitochondrial dysfunction after middle cerebral artery occlusion. We therefore propose that by inhibiting mitochondrial fission and attenuating mPTP opening, GK exerts neuroprotective effects that mitigate or prevent neuronal damage secondary to ischemic stroke.
Collapse
|
317
|
Zaric M, Drakulic D, Stojanovic IG, Mitrovic N, Grkovic I, Martinovic J. Regional-specific effects of cerebral ischemia/reperfusion and dehydroepiandrosterone on synaptic NMDAR/PSD-95 complex in male Wistar rats. Brain Res 2018; 1688:73-80. [PMID: 29577884 DOI: 10.1016/j.brainres.2018.03.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 10/17/2022]
Abstract
Excessive glutamate efflux and N-methyl-D-aspartate receptor (NMDAR) over-activation represent well-known hallmarks of cerebral ischemia/reperfusion (I/R) injury, still, expression of proteins involved in this aspect of I/R pathophysiology show inconsistent data. Neurosteroid dehydroepiandrosterone (DHEA) has been proposed as potent NMDAR modulator, but its influence on I/R-induced changes up to date remains questionable. Therefore, I/R-governed alteration of vesicular glutamate transporter 1 (vGluT1), synaptic NMDAR subunit composition, postsynaptic density protein 95 (PSD-95) and neuronal morphology alone or following DHEA treatment were examined. For that purpose, adult male Wistar rats were treated with a single dose of vehicle or DHEA (20 mg/kg i.p.) 4 h following sham operation or 15 min bilateral common carotid artery occlusion. Western blot was used for analyses of synaptic protein expressions in hippocampus and prefrontal cortex, while neuronal morphology was assessed using Nissl staining. Regional-specific postischemic changes were detected on protein level i.e. signs of neuronal damage in CA1 area was accompanied with hippocampal vGluT1, NR1, NR2B enhancement and PSD-95 decrement, while histological changes observed in layer III were associated with decreased NR1 subunit in prefrontal cortex. Under physiological conditions DHEA had no effect on protein and histological appearance, while in ischemic milieu it restored hippocampal PSD-95 and NR1 in prefrontal cortex to the control level. Along with intact neurons, ones characterized by morphology observed in I/R group were also present. Future studies involving NMDAR-related intracellular signaling and immunohistochemical analysis will reveal precise effects of I/R and DHEA treatment in selected brain regions.
Collapse
Affiliation(s)
- Marina Zaric
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Dunja Drakulic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Ivana Gusevac Stojanovic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Natasa Mitrovic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Ivana Grkovic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Jelena Martinovic
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
318
|
Park DJ, Shah FA, Koh PO. Quercetin attenuates neuronal cells damage in a middle cerebral artery occlusion animal model. J Vet Med Sci 2018; 80:676-683. [PMID: 29563391 PMCID: PMC5938200 DOI: 10.1292/jvms.17-0693] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cerebral ischemia is a neurological disorder with high mortality. Quercetin is a
flavonoid compound that is abundant in vegetables and fruits. It exerts anti-inflammatory
and anti-apoptotic effects. This study investigated the neuroprotective effects of
quercetin in focal cerebral ischemia. Male Sprague-Dawley rats were subjected to middle
cerebral artery occlusion (MCAO) to induce focal cerebral ischemia. Quercetin or vehicle
was injected 30 min before the onset of ischemia. A neurological function test, brain
edema measurement, and 2,3,5-triphenyltetrazolium chloride staining were performed to
elucidate the neuroprotective effects of quercetin. Western blot analysis was performed to
observe caspase-3 and poly ADP-ribose polymerase (PARP) protein expression. MCAO leads to
severe neuronal deficits and increases brain edema and infarct volume. However, quercetin
administration attenuated the MCAO-induced neuronal deficits and neuronal degeneration. We
observed increases in caspase-3 and PARP protein levels in MCAO-operated animals injected
with vehicle, whereas quercetin administration attenuated these increases in MCAO injury.
This study reveals the neuroprotective effect of quercetin in an MCAO-induced animal model
and demonstrates the regulation of caspase-3 and PARP expression by quercetin treatment.
These results suggest that quercetin exerts a neuroprotective effect through preventing
the MCAO-induced activation of apoptotic pathways affecting caspase-3 and PARP
expression.
Collapse
Affiliation(s)
- Dong-Ju Park
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Fawad-Ali Shah
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Phil-Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
319
|
Inhibition of mitochondrial permeability transition pore opening contributes to cannabinoid type 1 receptor agonist ACEA-induced neuroprotection. Neuropharmacology 2018; 135:211-222. [PMID: 29574098 DOI: 10.1016/j.neuropharm.2018.03.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 11/23/2022]
Abstract
Cannabinoid type 1 (CB1) receptor agonist arachidonyl-2-chloroethylamide (ACEA) induces neuroprotection against brain ischemia, and the mechanism, however, is still elusive. In this study, we used bilateral common carotid artery occlusion (BCCAO) in mice and oxygen-glucose deprivation (OGD) in primary cultured neurons to mimic brain ischemic injury, and hypothesized that cannabinoid CB1 receptor agonist ACEA protects ischemic neurons via inhibiting the opening of mitochondrial permeability transition pore (MPTP). In vivo, we found that BCCAO treatment reduced the neurological functions, increased the number of apoptotic neuronal cells and deteriorated the mitochondrial morphology in the ischemic brain tissue. And in vitro, we observed that OGD injury reduced cell viability, mitochondrial function and anti-oxidant SOD2 expression, increased lactate dehydrogenase (LDH), mitochondrial cytochrome C (Cyto C) and apoptosis-inducing factor (AIF) releases, elevated the cell apoptosis and mitochondrial superoxide level. And the CB1 receptor agonist ACEA significantly abolished the BCCAO and OGD-induced neuronal injury above. However, the MPTP opener atractyloside (Atr) markedly reversed the ACEA-induced neuroprotective effects, inhibited the mitochondrial Cyto C and AIF releases and relieved the mitochondrial swelling, but the MPTP inhibitor cyclosporin A (CsA) did not cause significant effects on the ACEA-induced neuroprotection above. These findings indicated that inhibition of MPTP opening may be involved in the cannabinoid CB1 receptor agonist ACEA-induced neuroprotection.
Collapse
|
320
|
Babenko VA, Silachev DN, Popkov VA, Zorova LD, Pevzner IB, Plotnikov EY, Sukhikh GT, Zorov DB. Miro1 Enhances Mitochondria Transfer from Multipotent Mesenchymal Stem Cells (MMSC) to Neural Cells and Improves the Efficacy of Cell Recovery. Molecules 2018; 23:molecules23030687. [PMID: 29562677 PMCID: PMC6017474 DOI: 10.3390/molecules23030687] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/13/2018] [Accepted: 03/17/2018] [Indexed: 01/08/2023] Open
Abstract
A recently discovered key role of reactive oxygen species (ROS) in mitochondrial traffic has opened a wide alley for studying the interactions between cells, including stem cells. Since its discovery in 2006, intercellular mitochondria transport has been intensively studied in different cellular models as a basis for cell therapy, since the potential of replacing malfunctioning organelles appears to be very promising. In this study, we explored the transfer of mitochondria from multipotent mesenchymal stem cells (MMSC) to neural cells and analyzed its efficacy under normal conditions and upon induction of mitochondrial damage. We found that mitochondria were transferred from the MMSC to astrocytes in a more efficient manner when the astrocytes were exposed to ischemic damage associated with elevated ROS levels. Such transport of mitochondria restored the bioenergetics of the recipient cells and stimulated their proliferation. The introduction of MMSC with overexpressed Miro1 in animals that had undergone an experimental stroke led to significantly improved recovery of neurological functions. Our data suggest that mitochondrial impairment in differentiated cells can be compensated by receiving healthy mitochondria from MMSC. We demonstrate a key role of Miro1, which promotes the mitochondrial transfer from MMSC and suggest that the genetic modification of stem cells can improve the therapies for the injured brain.
Collapse
Affiliation(s)
- Valentina A Babenko
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia.
| | - Denis N Silachev
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia.
| | - Vasily A Popkov
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia.
| | - Ljubava D Zorova
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia.
| | - Irina B Pevzner
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia.
| | - Egor Y Plotnikov
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia.
| | - Gennady T Sukhikh
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia.
- Department of obstetrics, gynecology, perinatology and reproduction, Sechenov First Moscow State Medical University, 119991 Moscow, Russia.
| | - Dmitry B Zorov
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia.
| |
Collapse
|
321
|
Abstract
Reactive oxygen species (ROS) are well known for their role in mediating both physiological and pathophysiological signal transduction. Enzymes and subcellular compartments that typically produce ROS are associated with metabolic regulation, and diseases associated with metabolic dysfunction may be influenced by changes in redox balance. In this review, we summarize the current literature surrounding ROS and their role in metabolic and inflammatory regulation, focusing on ROS signal transduction and its relationship to disease progression. In particular, we examine ROS production in compartments such as the cytoplasm, mitochondria, peroxisome, and endoplasmic reticulum and discuss how ROS influence metabolic processes such as proteasome function, autophagy, and general inflammatory signaling. We also summarize and highlight the role of ROS in the regulation metabolic/inflammatory diseases including atherosclerosis, diabetes mellitus, and stroke. In order to develop therapies that target oxidative signaling, it is vital to understand the balance ROS signaling plays in both physiology and pathophysiology, and how manipulation of this balance and the identity of the ROS may influence cellular and tissue homeostasis. An increased understanding of specific sources of ROS production and an appreciation for how ROS influence cellular metabolism may help guide us in the effort to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Daniel S Kikuchi
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Marina S Hernandes
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Qian Xu
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Kathy K Griendling
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA.
| |
Collapse
|
322
|
Anzell AR, Maizy R, Przyklenk K, Sanderson TH. Mitochondrial Quality Control and Disease: Insights into Ischemia-Reperfusion Injury. Mol Neurobiol 2018; 55:2547-2564. [PMID: 28401475 PMCID: PMC5636654 DOI: 10.1007/s12035-017-0503-9] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/20/2017] [Indexed: 12/28/2022]
Abstract
Mitochondria are key regulators of cell fate during disease. They control cell survival via the production of ATP that fuels cellular processes and, conversely, cell death via the induction of apoptosis through release of pro-apoptotic factors such as cytochrome C. Therefore, it is essential to have stringent quality control mechanisms to ensure a healthy mitochondrial network. Quality control mechanisms are largely regulated by mitochondrial dynamics and mitophagy. The processes of mitochondrial fission (division) and fusion allow for damaged mitochondria to be segregated and facilitate the equilibration of mitochondrial components such as DNA, proteins, and metabolites. The process of mitophagy are responsible for the degradation and recycling of damaged mitochondria. These mitochondrial quality control mechanisms have been well studied in chronic and acute pathologies such as Parkinson's disease, Alzheimer's disease, stroke, and acute myocardial infarction, but less is known about how these two processes interact and contribute to specific pathophysiologic states. To date, evidence for the role of mitochondrial quality control in acute and chronic disease is divergent and suggests that mitochondrial quality control processes can serve both survival and death functions depending on the disease state. This review aims to provide a synopsis of the molecular mechanisms involved in mitochondrial quality control, to summarize our current understanding of the complex role that mitochondrial quality control plays in the progression of acute vs chronic diseases and, finally, to speculate on the possibility that targeted manipulation of mitochondrial quality control mechanisms may be exploited for the rationale design of novel therapeutic interventions.
Collapse
Affiliation(s)
- Anthony R Anzell
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Rita Maizy
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Karin Przyklenk
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Thomas H Sanderson
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
323
|
Sanderson TH, Wider JM, Lee I, Reynolds CA, Liu J, Lepore B, Tousignant R, Bukowski MJ, Johnston H, Fite A, Raghunayakula S, Kamholz J, Grossman LI, Przyklenk K, Hüttemann M. Inhibitory modulation of cytochrome c oxidase activity with specific near-infrared light wavelengths attenuates brain ischemia/reperfusion injury. Sci Rep 2018; 8:3481. [PMID: 29472564 PMCID: PMC5823933 DOI: 10.1038/s41598-018-21869-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/13/2018] [Indexed: 12/17/2022] Open
Abstract
The interaction of light with biological tissue has been successfully utilized for multiple therapeutic purposes. Previous studies have suggested that near infrared light (NIR) enhances the activity of mitochondria by increasing cytochrome c oxidase (COX) activity, which we confirmed for 810 nm NIR. In contrast, scanning the NIR spectrum between 700 nm and 1000 nm revealed two NIR wavelengths (750 nm and 950 nm) that reduced the activity of isolated COX. COX-inhibitory wavelengths reduced mitochondrial respiration, reduced the mitochondrial membrane potential (ΔΨm), attenuated mitochondrial superoxide production, and attenuated neuronal death following oxygen glucose deprivation, whereas NIR that activates COX provided no benefit. We evaluated COX-inhibitory NIR as a potential therapy for cerebral reperfusion injury using a rat model of global brain ischemia. Untreated animals demonstrated an 86% loss of neurons in the CA1 hippocampus post-reperfusion whereas inhibitory NIR groups were robustly protected, with neuronal loss ranging from 11% to 35%. Moreover, neurologic function, assessed by radial arm maze performance, was preserved at control levels in rats treated with a combination of both COX-inhibitory NIR wavelengths. Taken together, our data suggest that COX-inhibitory NIR may be a viable non-pharmacologic and noninvasive therapy for the treatment of cerebral reperfusion injury.
Collapse
Affiliation(s)
- Thomas H Sanderson
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA. .,Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA. .,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA. .,Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| | - Joseph M Wider
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.,Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Icksoo Lee
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,College of Medicine, Dankook University, Cheonan-si, Chungcheongnam-do, 31116, Republic of Korea
| | - Christian A Reynolds
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Jenney Liu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Bradley Lepore
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Reneé Tousignant
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Melissa J Bukowski
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Hollie Johnston
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Alemu Fite
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Sarita Raghunayakula
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - John Kamholz
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Lawrence I Grossman
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Karin Przyklenk
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Maik Hüttemann
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA. .,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
324
|
Komnig D, Gertz K, Habib P, Nolte KW, Meyer T, Brockmann MA, Endres M, Rathkolb B, Hrabě de Angelis M, Schulz JB, Falkenburger BH, Reich A. Faim2 contributes to neuroprotection by erythropoietin in transient brain ischemia. J Neurochem 2018; 145:258-270. [PMID: 29315561 DOI: 10.1111/jnc.14296] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/22/2017] [Accepted: 12/23/2017] [Indexed: 11/28/2022]
Abstract
Delayed cell death in the penumbra region of acute ischemic stroke occurs through apoptotic mechanisms, making it amenable to therapeutic interventions. Fas/CD95 mediates apoptotic cell death in response to external stimuli. In mature neurons, Fas/CD95 signaling is modulated by Fas-apoptotic inhibitory molecule 2 (Faim2), which reduces cell death in animal models of stroke, meningitis, and Parkinson disease. Erythropoietin (EPO) has been studied as a therapeutic strategy in ischemic stroke. Erythropoietin stimulates the phosphatidylinositol-3 kinase/Akt (PI3K/Akt) pathway, which regulates Faim2 expression. Therefore, up-regulation of Faim2 may contribute to neuroprotection by EPO. Male Faim2-deficient mice (Faim2-/- ) and wild-type littermates (WT) were subjected to 30 min of middle cerebral artery occlusion (MCAo) followed by 72 h of reperfusion. EPO was applied before (30 min) and after (24 and 48 h) MCAo. In WT mice application of EPO at a low dose (5000 U/kg) significantly reduced stroke volume, whereas treatment with high dose (90 000 U/kg) did not. In Faim2-/- animals administration of low-dose EPO did not result in a significant reduction in stroke volume. Faim2 expression as measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR) increased after low-dose EPO but not with high dose. An extensive phenotyping including analysis of cerebral vessel architecture did not reveal confounding differences between the genotypes. In human post-mortem brain Faim2 displayed a differential expression in areas of penumbral ischemia. Faim2 up-regulation may contribute to the neuroprotective effects of low-dose erythropoietin in transient brain ischemia. The dose-dependency may explain mixed effects of erythropoietin observed in clinical stroke trials.
Collapse
Affiliation(s)
- Daniel Komnig
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Karen Gertz
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Pardes Habib
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Kay W Nolte
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Tareq Meyer
- Department of Diagnostic and Interventional Neuroradiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Endres
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany.,Excellence Cluster NeuroCure, Berlin, Germany.,German Center for Neurodegenerative Disease (DZNE), Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Ludwig-Maximilians-Universität München, Gene Center, Institute of Molecular Animal Breeding and Biotechnology, München, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, Freising-Weihenstephan, Germany
| | | | - Jörg B Schulz
- Department of Neurology, RWTH Aachen University, Aachen, Germany.,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Björn H Falkenburger
- Department of Neurology, RWTH Aachen University, Aachen, Germany.,JARA-BRAIN Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
325
|
Li C, Sun H, Xu G, McCarter KD, Li J, Mayhan WG. Mito-Tempo prevents nicotine-induced exacerbation of ischemic brain damage. J Appl Physiol (1985) 2018; 125:49-57. [PMID: 29420160 DOI: 10.1152/japplphysiol.01084.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Nicotine may contribute to the pathogenesis of cerebrovascular disease via the generation of reactive oxygen species (ROS). Overproduction of ROS leads to brain damage by intensifying postischemic inflammation. Our goal was to determine the effect of Mito-Tempo, a mitochondria-targeted antioxidant, on ischemic brain damage and postischemic inflammation during chronic exposure to nicotine. Male Sprague-Dawley rats were divided into four groups: control, nicotine, Mito-Tempo-treated control, and Mito-Tempo-treated nicotine. Nicotine (2 mg·kg-1·day-1) was administered via an osmotic minipump for 4 wk. Mito-Tempo (0.7 mg·kg-1·day-1 ip) was given for 7 days before cerebral ischemia. Transient focal cerebral ischemia was induced by occlusion of the middle cerebral artery for 2 h. Brain damage and inflammation were evaluated after 24 h of reperfusion by measuring infarct volume, expression of adhesion molecules, activity of matrix metalloproteinase, brain edema, microglial activation, and neutrophil infiltration. Nicotine exacerbated infarct volume and worsened neurological deficits. Nicotine did not alter baseline ICAM-1 expression, matrix metallopeptidase-2 activity, microglia activation, or neutrophil infiltration but increased these parameters after cerebral ischemia. Mito-Tempo did not have an effect in control rats but prevented the chronic nicotine-induced augmentation of ischemic brain damage and postischemic inflammation. We suggest that nicotine increases brain damage following cerebral ischemia via an increase in mitochondrial oxidative stress, which, in turn, contributes to postischemic inflammation. NEW & NOTEWORTHY Our findings have important implications for the understanding of mechanisms contributing to increased susceptibility of the brain to damage in smokers and users of nicotine-containing tobacco products.
Collapse
Affiliation(s)
- Chun Li
- Department of Cellular Biology and Anatomy, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Hong Sun
- Department of Cellular Biology and Anatomy, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Guodong Xu
- Department of Cellular Biology and Anatomy, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport , Shreveport, Louisiana.,Department of Neurology, Hebei General Hospital , Shijiazhuang, Hebei , China
| | - Kimberly D McCarter
- Department of Cellular Biology and Anatomy, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - Jiyu Li
- Department of Cellular Biology and Anatomy, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport , Shreveport, Louisiana
| | - William G Mayhan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, South Dakota
| |
Collapse
|
326
|
Gomez CR, Richards JG. Mitochondrial responses to anoxia exposure in red eared sliders (Trachemys scripta). Comp Biochem Physiol B Biochem Mol Biol 2018; 224:71-78. [PMID: 29402754 DOI: 10.1016/j.cbpb.2018.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 11/29/2022]
Abstract
When deprived oxygen, mitochondria from most vertebrates transform from the main site of ATP production to the dominant site of cellular ATP use due to the reverse functioning of the F1FO-ATPase (complex V). The anoxia-tolerant freshwater turtle Trachemys scripta however, has previously been shown to inhibit complex V activity in heart and brain in response to anoxia exposure, but the regulatory mechanism is unknown. To gain insight into the putative regulatory mechanisms underlying the anoxia-induced inhibition of complex V in T. scripta, we examined the effects of two weeks anoxia exposure at 4 °C on the mitochondrial proteome and candidate mechanisms that have been shown to regulate complex V in other organisms. In T. scripta, we confirmed that anoxia exposure resulted in a >80% inhibition of complex V in heart, brain and liver. Incubation of mitochondria with the nitric oxide donor, s-nitrosoglutathione, did not affect complex V activity despite showing the expected inhibition in mice. Proteomics analysis showed anoxia-induced decreases in three peripheral stalk subunits of complex V, possibly pointing to a unique site of regulation. Proteomics analysis also revealed differential expression of numerous enzymes involved with the electron transport system, the tricarboxylic acid cycle, as well as lipid and amino acid metabolism in response to anoxia exposure.
Collapse
Affiliation(s)
- Crisostomo R Gomez
- Department of Zoology, The University of British Columbia, 6270 University Blvd, Vancouver V6T 1Z4, British Columbia, Canada
| | - Jeffrey G Richards
- Department of Zoology, The University of British Columbia, 6270 University Blvd, Vancouver V6T 1Z4, British Columbia, Canada.
| |
Collapse
|
327
|
Gaignard P, Fréchou M, Liere P, Thérond P, Schumacher M, Slama A, Guennoun R. Sex differences in brain mitochondrial metabolism: influence of endogenous steroids and stroke. J Neuroendocrinol 2018. [PMID: 28650095 DOI: 10.1111/jne.12497] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Steroids are neuroprotective and a growing body of evidence indicates that mitochondria are a potential target of their effects. The mitochondria are the site of cellular energy synthesis, regulate oxidative stress and play a key role in cell death after brain injury and neurodegenerative diseases. After providing a summary of the literature on the general functions of mitochondria and the effects of sex steroid administrations on mitochondrial metabolism, we summarise and discuss our recent findings concerning sex differences in brain mitochondrial function under physiological and pathological conditions. To analyse the influence of endogenous sex steroids, the oxidative phosphorylation system, mitochondrial oxidative stress and brain steroid levels were compared between male and female mice, either intact or gonadectomised. The results obtained show that females have higher a mitochondrial respiration and lower oxidative stress compared to males and also that these differences were suppressed by ovariectomy but not orchidectomy. We have also shown that the decrease in brain mitochondrial respiration induced by ischaemia/reperfusion is different according to sex. In both sexes, treatment with progesterone reduced the ischaemia/reperfusion-induced mitochondrial alterations. Our findings indicate sex differences in brain mitochondrial function under physiological conditions, as well as after stroke, and identify mitochondria as a target of the neuroprotective properties of progesterone. Thus, it is necessary to investigate sex specificity in brain physiopathological mechanisms, especially when mitochondria impairment is involved.
Collapse
Affiliation(s)
- P Gaignard
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
- Biochemistry Laboratory, Bicêtre Hospital, Assistance-Publique Hôpitaux de Paris, Kremlin-Bicêtre, France
| | - M Fréchou
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - P Liere
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - P Thérond
- Biochemistry Laboratory, Bicêtre Hospital, Assistance-Publique Hôpitaux de Paris, Kremlin-Bicêtre, France
| | - M Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| | - A Slama
- Biochemistry Laboratory, Bicêtre Hospital, Assistance-Publique Hôpitaux de Paris, Kremlin-Bicêtre, France
| | - R Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, Kremlin-Bicêtre, France
| |
Collapse
|
328
|
Lv J, Hu W, Yang Z, Li T, Jiang S, Ma Z, Chen F, Yang Y. Focusing on claudin-5: A promising candidate in the regulation of BBB to treat ischemic stroke. Prog Neurobiol 2018; 161:79-96. [PMID: 29217457 DOI: 10.1016/j.pneurobio.2017.12.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/20/2017] [Accepted: 12/03/2017] [Indexed: 12/11/2022]
Abstract
Claudin-5 is a tight junction (TJ) protein in the blood-brain barrier (BBB) that has recently attracted increased attention. Numerous studies have demonstrated that claudin-5 regulates the integrity and permeability of the BBB. Increased claudin-5 expression plays a neuroprotective role in neurological diseases, particularly in cerebral ischemic stroke. Moreover, claudin-5 might be a potential marker for early hemorrhagic transformation detection in ischemic stroke. In light of the distinctive effects of claudin-5 on the nervous system, we present the elaborate network of roles that claudin-5 plays in ischemic stroke. In this review, we first introduce basic knowledge regarding the BBB and the claudin family, the characterization and regulation of claudin-5, and association between claudin-5 and other TJ proteins. Subsequently, we describe BBB dysfunction and neuron-specific drivers of pathogenesis of ischemic stroke, including inflammatory disequilibrium and oxidative stress. Furthermore, we summarize promising ischemic stroke treatments that target the BBB via claudin-5, including modified rt-PA therapy, pharmacotherapy, hormone treatment, receptor-targeted therapy, gene therapy, and physical therapy. This review highlights recent advances and provides a comprehensive summary of claudin-5 in the regulation of the BBB and may be helpful for drug design and clinical therapy for treatment of ischemic stroke.
Collapse
Affiliation(s)
- Jianjun Lv
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China; Department of Immunology, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Fulin Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
329
|
Jang DH, Khatri UG, Shortal BP, Kelly M, Hardy K, Lambert DS, Eckmann DM. Alterations in mitochondrial respiration and reactive oxygen species in patients poisoned with carbon monoxide treated with hyperbaric oxygen. Intensive Care Med Exp 2018; 6:4. [PMID: 29383459 PMCID: PMC5790762 DOI: 10.1186/s40635-018-0169-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/10/2018] [Indexed: 12/03/2022] Open
Abstract
Background Carbon monoxide (CO) poisoning is the leading cause of poisoning mortality and morbidity in the USA. Carboxyhemoglobin (COHb) levels are not predictive of severity or prognosis. At this time, the measurement of mitochondrial respiration may serve as a biomarker in CO poisoning. The primary objective of this study was to assess changes in mitochondrial function consisting of respiration and generation of reactive oxygen species (ROS) in peripheral blood mononuclear cells (PBMCs) obtained from patients with CO poisoning. Methods PBMCs from patients having confirmed CO exposure treated with hyperbaric oxygen or HBO (CO group) and healthy controls (control group) were analyzed with high-resolution respirometry. PBMCs were placed in a 2-ml chamber at a final concentration of 3–4 × 106 cells/ml to simultaneously obtain both respiration and hydrogen peroxide (H2O2) production. In the CO group, we performed measurements before and after patients underwent their first HBO treatment. Results We enrolled a total of 17 subjects, including 7 subjects with confirmed CO poisoning and 10 subjects in the control group. The CO group included five (71.4%) men and two (28.6%) women having a median COHb of 28%. There was a significant decrease in respiration as measured in pmol O2 × s− 1 × 10− 6 PBMCs in the CO group (pre-HBO) when compared to the control group: maximal respiration (18.4 ± 2.4 versus 35.4 ± 2.8, P < 0.001); uncoupled Complex I respiration (19.8 ± 1.8 versus 41.1 ± 3.8, P < 0.001); uncoupled Complex I + II respiration (32.3 ± 3.2 versus 58.3 ± 3.1, P < 0.001); Complex IV respiration (43.5 ± 2.9 versus 63.6 ± 6.31, P < 0.05). There were also similar differences measured in the CO group before and after HBO treatment with an overall increase in respiration present after treatment. We also determined the rate of H2O2 production simultaneously with the measurement of respiration. There was an overall significant increase in the H2O2 production in the CO group after HBO treatment when compared to prior HBO treatment and the control group. Conclusions In this study, PBMCs obtained from subjects with CO poisoning have an overall decrease in respiration (similar H2O2 production) when compared to controls. The inhibition of Complex IV respiration is from CO binding leading to a downstream decrease in respiration at other complexes. PBMCs obtained from CO-poisoned individuals immediately following initial HBO therapy displayed an overall increase in both respiration and H2O2 production. The study findings demonstrate that treatment with HBO resulted in improved cellular respiration but a higher H2O2 production. It is unclear if the increased production of H2O2 in HBO treatment is detrimental.
Collapse
Affiliation(s)
- David H Jang
- Department of Emergency Medicine, Division of Medical Toxicology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, John Morgan Building, 3620 Hamilton Walk, Philadelphia, 19104, PA, USA.
| | - Utsha G Khatri
- Department of Emergency Medicine, Division of Medical Toxicology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, John Morgan Building, 3620 Hamilton Walk, Philadelphia, 19104, PA, USA
| | - Brenna P Shortal
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Matthew Kelly
- Department of Emergency Medicine, Division of Medical Toxicology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, John Morgan Building, 3620 Hamilton Walk, Philadelphia, 19104, PA, USA.,Department of Emergency Medicine, Division of Hyperbaric and Undersea Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Kevin Hardy
- Department of Emergency Medicine, Division of Medical Toxicology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, John Morgan Building, 3620 Hamilton Walk, Philadelphia, 19104, PA, USA.,Department of Emergency Medicine, Division of Hyperbaric and Undersea Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - David S Lambert
- Department of Emergency Medicine, Division of Medical Toxicology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, John Morgan Building, 3620 Hamilton Walk, Philadelphia, 19104, PA, USA.,Department of Emergency Medicine, Division of Hyperbaric and Undersea Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - David M Eckmann
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA.,Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, 19104, PA, USA.,Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, USA.,Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, USA.,Cardiovascular Institute, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
330
|
Kumfu S, Charununtakorn ST, Jaiwongkam T, Chattipakorn N, Chattipakorn SC. Humanin Exerts Neuroprotection During Cardiac Ischemia-Reperfusion Injury. J Alzheimers Dis 2018; 61:1343-1353. [DOI: 10.3233/jad-170708] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sirinart Kumfu
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Physiology, Cardiac Electrophysiology Unit, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Savitree T. Charununtakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Physiology, Cardiac Electrophysiology Unit, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Thidarat Jaiwongkam
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Physiology, Cardiac Electrophysiology Unit, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Physiology, Cardiac Electrophysiology Unit, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C. Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
331
|
Akpinar E, Sener M, Cadirci E, Halici Z, Cinar I, Kok A. An investigation of postmortem urotensin II receptor levels in brain and kidney tissues in a rat model of cardiac ischemia. JOURNAL OF FORENSIC SCIENCE AND MEDICINE 2018. [DOI: 10.4103/jfsm.jfsm_75_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
332
|
Seo EH, Song GY, Namgung JH, Oh CS, Lee SH, Kim SH. Receptor for activated C kinase 1 in rats with ischemia-reperfusion injury: intravenous versus inhalation anaesthetic agents. Int J Med Sci 2018; 15:352-358. [PMID: 29511370 PMCID: PMC5835705 DOI: 10.7150/ijms.22591] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/12/2018] [Indexed: 01/04/2023] Open
Abstract
Background: The study examined the difference in the expression of the receptor for activated C kinase 1 (RACK1) between anaesthesia with propofol and isoflurane in rats with myocardial ischemia-reperfusion injury (IRI). Methods: Male Sprague-Dawley rats were studied. Anaesthesia was induced with xylazine 20 µg/g by intraperitoneal injection and maintained with propofol or isoflurane. Myocardial IRI was induced by ligating the left anterior descending artery for 1 hour. Reactive oxygen species (ROS), cardiomyocyte apoptosis, the expression of RACK1 and toll-like receptor 4 (TLR4), and the heart injury score were compared between the two groups. Results: Cardiomyocyte apoptosis with ROS was significantly lower in the propofol group than in the isoflurane group. The propofol group had significantly higher RACK1 expression and lower TLR4 expression, compared with the isoflurane group (RACK1, 1970.50 ± 120.50 vs. 1350.20 ± 250.30, p<0.05; TLR4, 980.50 ± 110.75 vs. 1275.50 ± 75.35, p<0.05). However, the heart injury scores in the two groups did not differ significantly (3.56 ± 0.29 vs. 4.33 ± 0.23 in the propofol and isoflurane groups, respectively, p=0.33). Conclusion: There were significant differences in inflammation and apoptosis, including the expression of RACK1 and TLR4, after myocardial IRI between the propofol and isoflurane groups. However, both groups had similar heart injury scores.
Collapse
Affiliation(s)
- Eun-Hye Seo
- BK21 Plus, Department of Cellular and Molecular Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Ga-Yun Song
- Department of Microbiology, Konkuk University School of Medicine, Seoul, Korea
| | - Ji Hyeon Namgung
- Department of Microbiology, Konkuk University School of Medicine, Seoul, Korea
| | - Chung-Sik Oh
- Department of Medicine, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Korea
| | - Seung Hyun Lee
- Department of Microbiology, Konkuk University School of Medicine, Seoul, Korea.,Department of Medicine, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Korea
| | - Seong-Hyop Kim
- Department of Medicine, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Korea.,Department of Anesthesiology and Pain medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea.,Department of Infection and Immunology, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
333
|
Singer M. Critical illness and flat batteries. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2017; 21:309. [PMID: 29297363 PMCID: PMC5751585 DOI: 10.1186/s13054-017-1913-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An exaggerated, dysregulated host response to insults such as infection (i.e. sepsis), trauma and ischaemia-reperfusion injury can result in multiple organ dysfunction and death. While the focus of research in this area has largely centred on inflammation and immunity, a crucial missing link is the precise identification of mechanisms at the organ level that cause this physiological-biochemical failure. Any hypothesis must reconcile this functional organ failure with minimal signs of cell death, availability of oxygen, and (often) minimal early local inflammatory cell infiltrate. These failed organs also retain the capacity to usually recover, even those that are poorly regenerative. A metabolic-bioenergetic shutdown, akin to hibernation or aestivation, is the most plausible explanation currently advanced. This shutdown appears driven by a perfect storm of compromised mitochondrial oxidative phosphorylation related to inhibition by excessive inflammatory mediators, direct oxidant stress, a tissue oxygen deficit in the unresuscitated phase, altered hormonal drive, and downregulation of genes encoding mitochondrial proteins. In addition, the efficiency of oxidative phosphorylation may be affected by a substrate shift towards fat metabolism and increased uncoupling. A lack of sufficient ATP provision to fuel normal metabolic processes will drive downregulation of metabolism, and thus cellular functionality. In turn, a decrease in metabolism will provide negative feedback to the mitochondrion, inducing a bioenergetic shutdown. Arguably, these processes may offer protection against a prolonged inflammatory hit by sparing the cell from initiation of death pathways, thereby explaining the lack of significant morphological change. A narrow line may exist between adaptation and maladaptation. This places a considerable challenge on any therapeutic modulation to provide benefit rather than harm.
Collapse
Affiliation(s)
- Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Cruciform Building, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
334
|
Withania coagulans Protects Striatum from Oxidative Damages Induced by Global Brain Ischemia in Rat. Jundishapur J Nat Pharm Prod 2017. [DOI: 10.5812/jjnpp.65051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
335
|
The Crosstalk between ROS and Autophagy in the Field of Transplantation Medicine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7120962. [PMID: 29410735 PMCID: PMC5749284 DOI: 10.1155/2017/7120962] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/21/2017] [Accepted: 10/08/2017] [Indexed: 12/17/2022]
Abstract
Many factors during the transplantation process influence posttransplant graft function and survival, including donor type and age, graft preservation methods (cold storage, machine perfusion), and ischemia-reperfusion injury. Successively, they will lead to cellular and molecular alterations that determine cell and ultimately organ fate. Oxidative stress and autophagy are implicated in posttransplant outcome since they are both affected by the stress responses triggered in each step (donor, preservation, and recipient) of the transplantation process. Furthermore, oxidative stress influences autophagy and vice versa. Interestingly, both processes have positive as well as negative effects on graft outcome, suggesting they are tightly linked during the transplantation process. In this review, we discuss the importance, regulation and crosstalk of oxidative signals, and autophagy in the field of transplantation medicine.
Collapse
|
336
|
Down-regulation of inflammatory signaling pathways despite up-regulation of Toll-like receptors; the effects of corticosteroid therapy in brain-dead kidney donors, a double-blind, randomized, controlled trial. Mol Immunol 2017; 94:36-44. [PMID: 29253747 DOI: 10.1016/j.molimm.2017.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND The brain death of a potential organ donor induces a systemic inflammatory response, resulting in inferior organ quality and function. Our study aimed to evaluate the effects of methylprednisolone (MPN) therapy on pattern recognition receptor (PRR) signaling in potential brain-dead (BD) kidney donors. MATERIAL AND METHODS To evaluate the effects of MPN therapy on PRR signaling in BD kidney donors we performed a prospective randomized treatment-versus-control study. Fifty-one potential kidney donors were randomly divided into three groups: brain-dead donors (BDDs) who received 15 mg/kg/d of methylprednisolone (group T1, n = 17), BDDs who received 15 mg/kg/d of MPN at the time of filling consent for kidney donation and 100 mg/2 h until kidney harvest (group T2, n = 17), and normal donors as controls n = 17. Gene expression for Toll-like receptors (TLRs) 1-9 and their signaling pathway molecules including MYD88, TRIF, NF-KB1, IRAK, IRF3, and IRF7, as well as the inflammatory cytokines RANTES, IL-1β, TNF-α, IL-6, CXCL8, IL-18, IFN-α, and IFN-β was determined by PCR array. Due to the crucial role of TLRs 2 and 4 in pattern recognition, surface expression of these molecules was analyzed by flow cytometry. Plasma levels of inflammatory cytokines were measured by immunoassay. Finally, serum creatinine and cystatin C were measured in 100 kidney recipients one week and one, three, and six months after transplant. RESULT Polymerase chain reaction (PCR) array gene expression revealed greater expression of TLRs and signaling molecules in group T1 than in the controls. Surface expression of TLRs 2 and 4 were significantly greater in group T2 than in group T1 (P < .05). Plasma concentrations of inflammatory cytokines were significantly greater in group T1 than in controls (P < .05). The recipients that received kidneys from group T1 had significantly higher levels of creatinine and cystatin C than the recipients of kidneys from both group T1 and controls (P<0.05). CONCLUSION Administration of MPN to BDDs at specified periods until kidney harvest resulted in less systemic inflammation in the BDDs and improved renal function in kidney graft recipients compared with common MPN therapy.
Collapse
|
337
|
Therapeutic Potential of Novel Twin Compounds Containing Tetramethylpyrazine and Carnitine Substructures in Experimental Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7191856. [PMID: 29387294 PMCID: PMC5745738 DOI: 10.1155/2017/7191856] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 07/16/2017] [Accepted: 08/13/2017] [Indexed: 02/07/2023]
Abstract
Although studies have seen dramatic advances in the understanding of the pathogenesis of stroke such as oxidative stress, inflammation, excitotoxicity, calcium overload and apoptosis, the delivery of stroke therapies is still a great challenge. In this study, we designed and synthesized a series of novel twin compounds containing tetramethylpyrazine and carnitine substructures and explored their therapeutic potential and mechanism in stroke-related neuronal injury. We first screened the neuroprotective effects of candidate compounds and found that among the tested compounds, LR134 and LR143 exhibited significant neuroprotection as evidenced by reducing cerebral infarct and edema, improving neurological function as well as blood-brain barrier integrity in rats after cerebral ischemia/reperfusion injury. We further demonstrated that the neuroprotective effects of compounds LR134 and LR143 were associated with the reduced inflammatory responses and NADPH oxidase- (NOX2-) mediated oxidative stress and the protection of mitochondria accompanied by the improvement of energy supply. In summary, this study provides direct evidence showing that the novel twin compounds containing tetramethylpyrazine and carnitine substructures have neuroprotective effects with multiple therapeutic targets, suggesting that modulation of these chemical structures may be an innovative therapeutic strategy for treating patients with stroke.
Collapse
|
338
|
Li X, Yang W, Jiang LH. Alteration in Intracellular Zn 2+ Homeostasis as a Result of TRPM2 Channel Activation Contributes to ROS-Induced Hippocampal Neuronal Death. Front Mol Neurosci 2017; 10:414. [PMID: 29311807 PMCID: PMC5732979 DOI: 10.3389/fnmol.2017.00414] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/29/2017] [Indexed: 12/23/2022] Open
Abstract
Transient receptor potential melastatin-related 2 (TRPM2) channel, a molecular sensor for reactive oxygen species (ROS), plays an important role in cognitive dysfunction associated with post-ischemia brain damage thought to result from ROS-induced TRPM2-dependent neuronal death during reperfusion. Emerging evidence further suggests that an alteration in the Zn2+ homeostasis is critical in ROS-induced TRPM2-dependent neuronal death. Here we applied genetic and pharmacological interventions to define the role of TRPM2 channel in ROS-induced neuronal death and explore the mechanisms contributing in the alteration in intracellular Zn2+ homeostasis in mouse hippocampal neurons. Exposure of neurons to 30–300 μM H2O2 for 2–24 h caused concentration/duration-dependent neuronal death, which was significantly suppressed, but not completely prevented, by TRPM2-knockout (TRPM2-KO) and pharmacological inhibition of the TRPM2 channel. H2O2-induced neuronal death was also attenuated by treatment with TPEN acting as a Zn2+ selective chelator. Single cell imaging demonstrated that H2O2 evoked a prominent increase in the intracellular Zn2+ concentration, which was completely prevented by TPEN as well as TRPM2-KO and inhibition of the TRPM2 channel. Furthermore, H2O2 induced lysosomal Zn2+ release and lysosomal dysfunction, and subsequent mitochondrial Zn2+ accumulation that provokes mitochondrial dysfunction and ROS generation. These H2O2-induced lysosomal/mitochondrial effects were prevented by TRPM2-KO or TPEN. Taken together, our results provide evidence to show that a dynamic alteration in the intracellular Zn2+ homeostasis as a result of activation of the TRPM2 channel contributes to ROS-induced hippocampal neuronal death.
Collapse
Affiliation(s)
- Xin Li
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Wei Yang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
339
|
Feng Y, Hu Y. Bone morphogenetic protein 9 serves a protective role in response to ischemic‑reperfusion in the brain by promoting ERK activation. Mol Med Rep 2017; 17:2845-2852. [PMID: 29257291 PMCID: PMC5783498 DOI: 10.3892/mmr.2017.8253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate the expression and function mechanism of bone morphogenetic protein 9 (BMP9) in cerebral ischemia-reperfusion (I/R) injuries in vivo and in vitro. A total of 40 Sprague-Dawley rats were randomly divided into four groups (n=10): i) Normal control; ii) sham surgery group, the procedure without occlusion; iii) I/R group, right middle cerebral artery occlusion (MCAO) followed by reperfusion; and iv) adenoviral vector (Ad)-BMP9 + I/R group, Ad-BMP9 intracerebroventricular injection was performed 2 days prior to MCAO. Neurological deficit score and infarct volume were measured at 24 h following reperfusion. To further test the mechanism of BMP9, astrocytes were isolated and treated with Ad-BMP9, Ad-BMP9 + extracellular signal-regulated kinase (ERK) inhibitor PD098059, Ad-BMP9 + c-Jun N-terminal kinase inhibitor SP600125 and Ad-BMP9 + p38 inhibitor SB203580 for 24 h, followed by undergoing oxygen-glucose deprivation and reoxygenation (OGD/R) treatment. Cell viability and death were assessed by 3-(4,5-dimethylthiazol-2yl)-5-(3-carboxymethoxyphenyl)-(4-sulfophenyl)-2H-tetrazolium and lactate dehydrogenase release, respectively. Gene expression was determined by quantitative polymerase chain reaction and western blotting. BMP9 was identified to be upregulated at mRNA and protein levels in cerebral I/R animal and cell models. BMP9 pretreatment significantly reduced the neurological score and infarct volume compared with I/R rats. In astrocytes, overexpression of BMP9 significantly decreased cell death and improved cell viability, an effect which may be mediated by the ERK signaling pathway, as ERK was activated by BMP9 and the use of PD098059 partially reversed the protective effect of BMP9. Pretreatment with BMP-9 may be a promising treatment option for prevention of cerebral I/R injuries.
Collapse
Affiliation(s)
- Yinling Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yida Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
340
|
Liu CB, Qu GB, Cao MX, Liang Y, Hu LG, Shi JB, Cai Y, Jiang GB. Distinct toxicological characteristics and mechanisms of Hg 2+ and MeHg in Tetrahymena under low concentration exposure. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 193:152-159. [PMID: 29096088 DOI: 10.1016/j.aquatox.2017.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 06/07/2023]
Abstract
Inorganic divalent mercury complexes (Hg2+) and monomethylmercury complexes (MeHg) are the main mercury species in aquatic systems and their toxicity to aquatic organisms is of great concern. Tetrahymena is a type of unicellular eukaryotic protozoa located at the bottom of food chain that plays a fundamental role in the biomagnification of mercury. In this work, the dynamic accumulation properties, toxicological characteristics and mechanisms of Hg2+ and MeHg in five Tetrahymena species were evaluated in detail. The results showed that both Hg2+ and MeHg were ingested and exhibited inhibitory effects on the proliferation or survival of Tetrahymena species. However, the ingestion rate of MeHg was significantly higher than that of Hg2+. The mechanisms responsible for the toxicity of MeHg and Hg2+ were different, although both chemicals altered mitochondrial membrane potential (MMP). MeHg disrupted the integrity of membranes while Hg2+ had detrimental effects on Tetrahymena as a result of the increased generation of reactive oxygen species (ROS). In addition, the five Tetrahymena species showed different capacities in accumulating Hg2+ and MeHg, with T. corlissi exhibiting the highest accumulations. The study also found significant growth-promoting effect on T. corlissi under low concentration exposure (0.003 and 0.01μg Hg/mL (15 and 50nM)), suggesting different effect and mechanism that should be more closely examined when assessing the bioaccumulation and toxicity of mercury in aquatic ecosystems.
Collapse
Affiliation(s)
- Cheng-Bin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guang-Bo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng-Xi Cao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Yong Liang
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Li-Gang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jian-Bo Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Environment and Health, Jianghan University, Wuhan 430056, China.
| | - Yong Cai
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Gui-Bin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
341
|
Zhang CY, Sun XY, Ouyang JM, Gui BS. Diethyl citrate and sodium citrate reduce the cytotoxic effects of nanosized hydroxyapatite crystals on mouse vascular smooth muscle cells. Int J Nanomedicine 2017; 12:8511-8525. [PMID: 29238189 PMCID: PMC5713691 DOI: 10.2147/ijn.s145386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE This study aimed to investigate the damage mechanism of nanosized hydroxyapatite (nano-HAp) on mouse aortic smooth muscle cells (MOVASs) and the injury-inhibiting effects of diethyl citrate (Et2Cit) and sodium citrate (Na3Cit) to develop new drugs that can simultaneously induce anticoagulation and inhibit vascular calcification. METHODS The change in cell viability was evaluated using a cell proliferation assay kit, and the amount of lactate dehydrogenase (LDH) released was measured using an LDH kit. Intracellular reactive oxygen species (ROS) and mitochondrial damage were detected by DCFH-DA staining and JC-1 staining. Cell apoptosis and necrosis were detected by Annexin V staining. Intracellular calcium concentration and lysosomal integrity were measured using Fluo-4/AM and acridine orange, respectively. RESULTS Nano-HAp decreased cell viability and damaged the cell membrane, resulting in the release of a large amount of LDH. Nano-HAp entered the cells and damaged the mitochondria, and then induced cell apoptosis by producing a large amount of ROS. In addition, nano-HAp increased the intracellular Ca2+ concentration, leading to lysosomal rupture and cell necrosis. On addition of the anticoagulant Et2Cit or Na3Cit, cell viability and mitochondrial membrane potential increased, whereas the amount of LDH released, ROS, and apoptosis rate decreased. Et2 Cit and Na3Cit could also chelate with Ca+ to inhibit the intracellular Ca2+ elevations induced by nano-HAp, prevent lysosomal rupture, and reduce cell necrosis. High concentrations of Et2Cit and Na3Cit exhibited strong inhibitory effects. The inhibitory capacity of Na3Cit was stronger than that of Et2Cit at similar concentrations. CONCLUSION Both Et2Cit and Na3Cit significantly reduced the cytotoxicity of nano-HAp on MOVASs and inhibited the apoptosis and necrosis induced by nano-HAp crystals. The chelating function of citrate resulted in both anticoagulation and binding to HAp. Et2Cit and Na3Cit may play a role as anticoagulants in reducing injury to the vascular wall caused by nano-HAp.
Collapse
Affiliation(s)
- Chong-Yu Zhang
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou
| | - Xin-Yuan Sun
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou
| | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, Jinan University, Guangzhou
| | - Bao-Song Gui
- Department of Nephrology, The Second Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| |
Collapse
|
342
|
Akinmoladun AC, Saliu IO, Olowookere BD, Ojo OB, Olaleye MT, Farombi EO, Akindahunsi AA. Improvement of 2-Vessel Occlusion Cerebral Ischaemia/Reperfusion-Induced Corticostriatal Electrolyte and Redox Imbalance, Lactic Acidosis and Modified Acetylcholinesterase Activity by Kolaviron Correlates with Reduction in Neurobehavioural Deficits. Ann Neurosci 2017; 25:53-62. [PMID: 29887685 DOI: 10.1159/000484517] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/19/2017] [Indexed: 01/30/2023] Open
Abstract
Background Disruption of electrolyte, redox and neurochemical homeostasis alongside cellular energy crisis is a hallmark of cerebral ischaemia and reperfusion injury. Purpose This study investigated the effect of kolaviron (KV) on cortical and striatal cation imbalance, oxidative stress and neurochemical disturbances as well as neurobehavioural deficits in animals subjected to bilateral common carotid artery occlusion (BCCAO)-induced ischaemia/reperfusion injury. Methods KV was administered at a dose of 100 or 200 mg/kg to male Wistar rats 1 h before a 30 min BCCAO/4 h reperfusion (I/R). This was followed by neurobehavioral assessment and biochemical evaluations of cation levels, oxidative stress indicators, lactate dehydrogenase activity and acetylcholinesterase (AChE) activity in the brain of animals. Conclusion KV significantly restored altered cortical and striatal Ca2+, Na+, K+ and Mg2+ levels, ameliorated redox imbalance, lactic acidosis and modified AChE activity caused by I/R injury. The favourable neurobehavioural effects of KV correlated with biochemical outcomes. The pharmacological potential of KV in the treatment and management of ischemic stroke and allied pathological conditions via multiple targets (neurotransmitter metabolism, bioenergetic failure and ionic homeostasis) is highlighted by the study.
Collapse
Affiliation(s)
- Afolabi Clement Akinmoladun
- Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
| | - Ibrahim Olabayode Saliu
- Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
| | - Boyede Dele Olowookere
- Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
| | - Olubukola Benedicta Ojo
- Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
| | - Mary Tolulope Olaleye
- Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
| | - Ebenezer Olatunde Farombi
- Drug Metabolism and Molecular Toxicology Unit, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Afolabi Akintunde Akindahunsi
- Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, The Federal University of Technology, Akure, Nigeria
| |
Collapse
|
343
|
Ginseng Rh2 protects endometrial cells from oxygen glucose deprivation/re-oxygenation. Oncotarget 2017; 8:105703-105713. [PMID: 29285285 PMCID: PMC5739672 DOI: 10.18632/oncotarget.22390] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/27/2017] [Indexed: 01/22/2023] Open
Abstract
In this study, oxygen glucose deprivation/re-oxygenation (OGDR) was applied to cultured endometrial cells to mimic ischemic-reperfusion injuries. We also tested the potential effect of Ginseng Rh2 (GRh2) against the process. In established T-HESC human endometrial cells and primary murine endometrial cells, GRh2 largely inhibited OGDR-induced viability reduction and cell death. Remarkably, OGDR induced programmed necrosis in the endometrial cells, evidenced by cyclophilin D-p53-adenine nucleotide translocator 1 (ANT-1) mitochondrial association, mitochondrial depolarization, reactive oxygen species production, and lactate dehydrogenase release. Notably, such effects by OGDR were largely attenuated with co-treatment of GRh2. Further, cyclophilin D inhibition or knockdown also protected endometrial cells from OGDR. On the other hand, forced over-expression of cyclophilin D facilitated OGDR-induced T-HESC cell necrosis, which was dramatically inhibited by GRh2. Together, GRh2 protects endometrial cells from OGDR possibly via inhibiting CypD-dependent programmed necrosis pathway.
Collapse
|
344
|
Ma L, Liu JY, Dong JX, Xiao Q, Zhao J, Jiang FL. Toxicity of Pb 2+ on rat liver mitochondria induced by oxidative stress and mitochondrial permeability transition. Toxicol Res (Camb) 2017; 6:822-830. [PMID: 30090545 PMCID: PMC6062357 DOI: 10.1039/c7tx00204a] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/24/2017] [Indexed: 12/26/2022] Open
Abstract
Pb2+ exposure in humans occurs mainly through air inhalation, food and water uptake which has been shown to be generally associated with numerous body functions such as the central and peripheral nervous systems, the red blood cells, the kidneys and the liver. It has been reported that the liver is the storage site and an important primary target in Pb2+ toxicity, and the hepatotoxicity of Pb2+ could be resulted from the impairment of the liver mitochondria. In this study, several mitochondrial dysfunctions following the addition of Pb2+ (10-160 μM) were investigated. We found that Pb2+ inhibited the enzyme activities of mitochondrial respiratory complexes and complex III was the major source of Pb2+-induced significant reactive oxygen species (ROS) formation. As a consequence, our results showed that Pb2+ induced significant progress in mitochondrial lipid peroxidation, adenosine triphosphate (ATP) consumption and glutathione (GSH) oxidation. On the other hand, Pb2+ induced marked changes in mitochondrial permeability transition (MPT) accompanied by mitochondrial swelling, mitochondrial membrane potential collapse, mitochondrial membrane fluidity decrease and cytochrome c (Cyt c) release. Additionally, several mitochondrial MPT inhibitors and chelators were utilized to determine the possible interaction sites of Pb2+ on mitochondria. In general, our data supported that the Pb2+-induced liver toxicity was a result of the disruptive effect on the mitochondrial respiratory complexes. This disruptive effect caused oxidative stress and MPT, which led to mitochondrial dysfunctions and even cell death signalling via mitochondrial permeability transition pore (MPTP) opening and Cyt c release.
Collapse
Affiliation(s)
- Long Ma
- State Key Laboratory of Virology & Key Laboratory of Analytical Chemistry for Biology and Medicine (MOE) , College of Chemistry and Molecular Sciences , Wuhan University , Wuhan 430072 , P. R. China . ; ; Tel: +86-27-68756667
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmaceutical Sciences , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Jun-Yi Liu
- The Bryn Mawr School , Baltimore , MD 21210 , USA
| | - Jia-Xin Dong
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmaceutical Sciences , Guangxi Normal University , Guilin 541004 , P. R. China
| | - Qi Xiao
- College of Chemistry and Material Science , Guangxi Teachers Education University , Nanning 530001 , P. R. China
| | - Jie Zhao
- State Key Laboratory of Virology & Key Laboratory of Analytical Chemistry for Biology and Medicine (MOE) , College of Chemistry and Molecular Sciences , Wuhan University , Wuhan 430072 , P. R. China . ; ; Tel: +86-27-68756667
| | - Feng-Lei Jiang
- State Key Laboratory of Virology & Key Laboratory of Analytical Chemistry for Biology and Medicine (MOE) , College of Chemistry and Molecular Sciences , Wuhan University , Wuhan 430072 , P. R. China . ; ; Tel: +86-27-68756667
| |
Collapse
|
345
|
Jones AR, Frazier SK. Consequences of Transfusing Blood Components in Patients With Trauma: A Conceptual Model. Crit Care Nurse 2017; 37:18-30. [PMID: 28365647 DOI: 10.4037/ccn2017965] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Transfusion of blood components is often required in resuscitation of patients with major trauma. Packed red blood cells and platelets break down and undergo chemical changes during storage (known as the storage lesion) that lead to an inflammatory response once the blood components are transfused to patients. Although some evidence supports a detrimental association between transfusion and a patient's outcome, the mechanisms connecting transfusion of stored components to outcomes remain unclear. The purpose of this review is to provide critical care nurses with a conceptual model to facilitate understanding of the relationship between the storage lesion and patients' outcomes after trauma; outcomes related to trauma, hemorrhage, and blood component transfusion are grouped according to those occurring in the short-term (≤30 days) and the long-term (>30 days). Complete understanding of these clinical implications is critical for practitioners in evaluating and treating patients given transfusions after traumatic injury.
Collapse
Affiliation(s)
- Allison R Jones
- Allison R. Jones is an assistant professor, Department of Acute, Chronic, and Continuing Care, School of Nursing, University of Alabama, Birmingham, Alabama. She has a clinical background in emergency and trauma nursing. In research, she focuses on the consequences of blood component storage and transfusion, with particular interest in transfusion after trauma. .,Susan K. Frazier is the director of the PhD program, a codirector of the RICH Heart Program, and an associate professor, College of Nursing, University of Kentucky, Lexington, Kentucky. Her research focuses on cardiopulmonary interactions in a variety of critically ill patients, including patients with acute heart failure, acute decompensated heart failure, acute respiratory distress syndrome, chronic obstructive pulmonary disease, and multiple trauma.
| | - Susan K Frazier
- Allison R. Jones is an assistant professor, Department of Acute, Chronic, and Continuing Care, School of Nursing, University of Alabama, Birmingham, Alabama. She has a clinical background in emergency and trauma nursing. In research, she focuses on the consequences of blood component storage and transfusion, with particular interest in transfusion after trauma.,Susan K. Frazier is the director of the PhD program, a codirector of the RICH Heart Program, and an associate professor, College of Nursing, University of Kentucky, Lexington, Kentucky. Her research focuses on cardiopulmonary interactions in a variety of critically ill patients, including patients with acute heart failure, acute decompensated heart failure, acute respiratory distress syndrome, chronic obstructive pulmonary disease, and multiple trauma
| |
Collapse
|
346
|
Chen H, Song Z, Ying S, Yang X, Wu W, Tan Q, Ju X, Wu W, Zhang X, Qu J, Wang Y. Myeloid differentiation protein 2 induced retinal ischemia reperfusion injury via upregulation of ROS through a TLR4-NOX4 pathway. Toxicol Lett 2017; 282:109-120. [PMID: 29111459 DOI: 10.1016/j.toxlet.2017.10.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/25/2017] [Accepted: 10/26/2017] [Indexed: 10/18/2022]
Abstract
Retinal ischemia reperfusion (I/R) injury is common in many ophthalmic diseases. Recent studies have shown that toll-like receptor 4 (TLR4) is involved in ischemic retinal injury. Activation of TLRs requires specific accessory proteins such as myeloid differentiation protein 2 (MD2), which facilitate in ligand responsiveness. Therefore, inhibiting MD2 may be a novel approach to modulate TLR4 signaling and deleterious downstream effects in ischemic retinal injury. We used human Müller MIO-M1 cells treated with tert-butyl hydroperoxide (TBHP) to establish an in vitro I/R model of oxidative injury and tested the therapeutic effect of inhibiting MD2. Furthermore, we inhibited MD2 in a mouse model of retinal I/R injury and confirmed the results using MD2 knockout mice. Our studies show that pharmacological inhibition of MD2 prevented TBHP-induced reactive oxygen species (ROS) generation, inflammation and subsequent apoptosis in Müller cells. We also show that retinal I/R injury in mice induced functional deficits, increased ROS levels, inflammation and apoptosis. These pathological changes were not observed in MD2 knockout mice and attenuated when MD2 was inhibited in wildtype mice. In addition, we discovered that the mechanism of these therapeutic effects involved regulation of NADPH oxidase 4 (NOX4)-MD2-TLR4 complex formation. This study provides evidence that MD2 plays a key role in the pathogenesis of retinal I/R damage by participating in TLR4-NOX4 complex formation and elaboration of oxidative and inflammatory damage. Hence, inhibition of MD2 may reduce TLR-dependent damage during retinal I/R injury.
Collapse
Affiliation(s)
- Huaicheng Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zongming Song
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Shilong Ying
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xi Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Wu
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiufan Tan
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xin Ju
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wencan Wu
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xin Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia Qu
- The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
347
|
The UCP2-866G/A Polymorphism Could be Considered as a Genetic Marker of Different Functional Prognosis in Ischemic Stroke After Recanalization. Neuromolecular Med 2017; 19:571-578. [PMID: 29043564 DOI: 10.1007/s12017-017-8470-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022]
Abstract
Recent studies based on experimental animal models of stroke have suggested that uncoupling protein 2 (UCP2), an inner mitochondrial membrane protein that is thought to regulate energy metabolism and reduce reactive oxygen species generation, provides protection against reperfusion damage. We aimed to investigate whether -866G/A polymorphism in the promoter of the UCP2 gene, which enhances its transcriptional activity, is associated with functional prognosis in patients with embolic ischemic stroke after early recanalization. We investigate a hospital-based prospective cohort of patients with acute ischemic stroke due to occlusion of the middle cerebral artery diagnosed by transcranial Doppler who obtained a partial/complete recanalization 24 h after administration of intravenous thrombolysis. The main end point of the study was functional independence defined as modified Rankin Scale 0-2 on day 90. A total of 80 patients were enrolled. The UCP2-866G/A polymorphism was determined by polymerase chain reaction-restriction fragment length polymorphism technique (14 genotype A/A (18%), 45 genotype A/G (56%) and 21 genotype G/G (26%). The percentage of patients with good functional outcome at 3 months was significantly higher in patients harboring the A/A genotype than in those with A/G or G/G genotypes (85 vs 41%, p = 0.01). The A/A genotype was found to be an independent marker of good prognosis after adjustment for secondary variables (age, sex, glucose level, NIHSS score at baseline, complete recanalization and early neurological improvement) in a logistic regression analysis (OR 0.05, 95% CI 0.01-0.48, p = 0.01). Our results suggest that the AA genotype of UCP2-866 may predict a better functional outcome in ischemic stroke after recanalization of proximal MCA occlusion.
Collapse
|
348
|
Leiva-Salcedo E, Riquelme D, Cerda O, Stutzin A. TRPM4 activation by chemically- and oxygen deprivation-induced ischemia and reperfusion triggers neuronal death. Channels (Austin) 2017; 11:624-635. [PMID: 28876976 DOI: 10.1080/19336950.2017.1375072] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cerebral ischemia-reperfusion injury triggers a deleterious process ending in neuronal death. This process has two components, a glutamate-dependent and a glutamate-independent mechanism. In the glutamate-independent mechanism, neurons undergo a slow depolarization eventually leading to neuronal death. However, little is known about the molecules that take part in this process. Here we show by using mice cortical neurons in culture and ischemia-reperfusion protocols that TRPM4 is fundamental for the glutamate-independent neuronal damage. Thus, by blocking excitotoxicity, we reveal a slow activating, glibenclamide- and 9-phenanthrol-sensitive current, which is activated within 5 min upon ischemia-reperfusion onset. TRPM4 shRNA-based silenced neurons show a reduced ischemia-reperfusion induced current and depolarization. Neurons were protected from neuronal death up to 3 hours after the ischemia-reperfusion challenge. The activation of TRPM4 during ischemia-reperfusion injury involves the increase in both, intracellular calcium and H2O2, which may act together to produce a sustained activation of the channel.
Collapse
Affiliation(s)
- Elías Leiva-Salcedo
- a Departamento de Biología , Facultad de Química y Biología, Universidad de Santiago de Chile , Santiago , Chile
| | - Denise Riquelme
- a Departamento de Biología , Facultad de Química y Biología, Universidad de Santiago de Chile , Santiago , Chile
| | - Oscar Cerda
- b Programa de Biología Celular y Molecular , Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile.,c Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Chile , Santiago , Chile
| | - Andrés Stutzin
- d Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| |
Collapse
|
349
|
RTN1-C mediates cerebral ischemia/reperfusion injury via ER stress and mitochondria-associated apoptosis pathways. Cell Death Dis 2017; 8:e3080. [PMID: 28981095 PMCID: PMC5680587 DOI: 10.1038/cddis.2017.465] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/27/2017] [Accepted: 08/17/2017] [Indexed: 01/15/2023]
Abstract
The reticulon family has been found to induce apoptosis, inhibit axon regeneration and regulate protein trafficking. However, little is known about the mechanisms of how reticulon proteins are involved in neuronal death-promoting processes during ischemia. Here, we report that the expression of Reticulon Protein 1-C (RTN1-C) was associated with the progression of cerebral ischemia/reperfusion (I/R) injury. Using a combination of rat middle cerebral artery occlusion (MCAO) stroke and oxygen-glucose deprivation followed by reoxygenation (OGD/R) models, we determined that the expression of RTN1-C was significantly increased during cerebral ischemic/reperfusion. RTN1-C overexpression induced apoptosis and increased the cell vulnerability to ischemic injury, whereas RTN1-C knockdown reversed ischemia-induced apoptosis and attenuated the vulnerability of OGD/R-treated neural cells. Mechanistically, we demonstrated that RTN1-C mediated OGD/R-induced apoptosis through ER stress and mitochondria-associated pathways. RTN1-C interacted with Bcl-xL and increased its localization in the ER, thus reducing the anti-apoptotic activity of Bcl-xL. Most importantly, knockdown of Rtn1-c expression in vivo attenuated apoptosis in MCAO rats and reduced the extent of I/R-induced brain injury, as assessed by infarct volume and neurological score. Collectively, these data support for the first time that RTN1-C may represent a novel candidate for therapies against cerebral ischemia/reperfusion injury.
Collapse
|
350
|
Li J, Zhang Q, Li S, Dai W, Feng J, Wu L, Liu T, Chen K, Xia Y, Lu J, Zhou Y, Fan X, Guo C. The natural product fucoidan ameliorates hepatic ischemia-reperfusion injury in mice. Biomed Pharmacother 2017; 94:687-696. [PMID: 28797984 DOI: 10.1016/j.biopha.2017.07.109] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/08/2017] [Accepted: 07/24/2017] [Indexed: 12/29/2022] Open
Abstract
Fucoidan is a sulfated polysaccharide based predominantly on l-fucose, and has several biologic functions. Reactive oxygen species-mediated apoptosis and autophagy and release of related inflammatory factors have important roles in hepatic ischemia-reperfusion injury (IR). Here, the effect of fucoidan on hepatic IR was investigated. Mice were randomized into sham, IR, and fucoidan (20, 40mg/kg for 14days) groups. Samples were collected to assess biochemical indicators, hepatocyte damage and levels of proteins related to signaling pathways at different time points. Fucoidan had no effect on normal liver tissue, but inhibited the increases in alanine aminotransferase, aspartate transaminase, inflammatory factors, and the hepatocyte damage caused by IR. Also, apoptosis and autophagy via the activated JAK2/STAT1 pathway were attenuated by fucoidan to protect against hepatic injury. In conclusion, fucoidan ameliorates hepatic IR injury in mice via JAK2/STAT1-mediated apoptosis and autophagy. Inhibition of this pathway may be associated with reduced release of related inflammatory cytokines, especially interferon-γ.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - QingHui Zhang
- Department of Clinical Laboratory, Kunshan First People's Hospital Affiliated to Jiangsu University, 215300, Kunshan, JiangSu, China
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yujing Xia
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaoming Fan
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Jinshan, Shanghai 201508, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|