1
|
Joseph JP, Kumar T, Ramteke NS, Chatterjee K, Nandi D. High intracellular calcium amounts inhibit activation-induced proliferation of mouse T cells: Tert-butyl hydroquinone as an additive enhancer of intracellular calcium. Int Immunopharmacol 2024; 143:113501. [PMID: 39488036 DOI: 10.1016/j.intimp.2024.113501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/29/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
Optimal T cell activation is critical to orchestrate adaptive immune responses. Calcium is critical for T cell activation and integrates signaling pathways necessary to activate key transcription factors. In fact, patients with calcium channelopathies are immunodeficient. Here, we investigated the effects of different concentrations of intracellular calcium on activation of mouse T cells. High intracellular calcium amounts inhibited in vitro T cell proliferation as evidenced by a decreased cell cycling-to-hypodiploidy ratio in two models of activation: the combination of phorbol 12-myristate 13-acetate (PMA) and Ionomycin (an ionophore)/Thapsigargin (a SERCA inhibitor) or plate bound anti-CD3 and anti-CD28. High intracellular calcium amounts increased the production of reactive oxygen species (ROS) in T cells activated with PMA and Ionomycin and scavenging excess ROS using N-acetyl cysteine (NAC) rescued the decrease in cycling-to-hypodiploidy ratio. To test the universality of our observations, we studied the effects of tert-Butylhydroquinone (tBHQ), a SERCA inhibitor and Nrf2 activator. tBHQ alone did not increase intracellular calcium amounts but the intracellular calcium amounts increased when tBHQ was used in combination with PMA. Also, tBHQ inhibited T cell activation in a dose-dependent manner in both in vitro models of T cell activation. Importantly, intraperitoneal injection of tBHQ ameliorated Dextran Sodium Sulfate (DSS)-induced colitis in mice as evidenced by rescue of colon length shortening and lower disease activity index. Overall, this study identifies high calcium amounts as a potential target to lower T cell activation. The implications of these observations are discussed in the context of calcium modulating drugs that are used to treat various diseases.
Collapse
Affiliation(s)
- Joel P Joseph
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | - Tanisha Kumar
- Undergraduate Program, Indian Institute of Science, Bengaluru, India
| | - Nikita S Ramteke
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India
| | - Kaushik Chatterjee
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India; Department of Materials Engineering, Indian Institute of Science, Bengaluru, India
| | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
2
|
Triandafilova G, Smirnova G, Krasnykh O, Boteva A, Oktyabrsky O. Antimicrobial and Antioxidant Activity of Some Nitrogen-Containing Heterocycles and Their Acyclic Analogues. Indian J Microbiol 2024; 64:482-491. [PMID: 39011024 PMCID: PMC11246309 DOI: 10.1007/s12088-023-01158-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/17/2023] [Indexed: 07/17/2024] Open
Abstract
We investigated antimicrobial and antioxidant activity of nitrogen-containing heterocycles and their acyclic analogues, some of which can be considered as promising in terms of biological activity. Based on structure, 26 tested compounds were divided into 4 groups. In the test with 2,2-diphenyl-1-picrylhydrazyl (DPPH), the compounds of the group 2 had the highest radical-binding activity (RBA) (53-78%), while those of group 3 had the lowest values (1.5-5.2%). In oxygen radical absorbance capacity assay, all compounds from groups 1, 2 and 3 showed high RBA: 44-94% at 50 µM. The highest bacteriostatic activity against Escherichia coli was found for four compounds in group 2 (MIC = 0.25-1 mM) and low bacteriostatic activity for group 3 (MIC > 4 mM). Some relationships between the structure of compounds and the values of the MIC are revealed. It was also found that four substances from different groups had the ability to inhibit the formation of colonies in E. coli from 1.3 to 5.7 times. Four compounds reduced specific biofilm formation by 40-60%. The tested substances did not induce the expression of the sulA gene controlled by the SOS system, which indicates the lack of genotoxic activity. None of the tested compounds had pro-oxidant activity. This was shown by both the absence of production hydrogen peroxide in a bacteria-free medium and inability to induce expression of the katG gene encoding HPI catalase in growing E. coli. The data obtained could be useful in the development of new drugs.
Collapse
Affiliation(s)
- Galina Triandafilova
- Laboratory of Physiology and Genetics of Microorganisms, Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Russian Academy of Sciences, Perm, Russia
- Scientific and Educational Center for Applied Chemical and Biological Research, Perm National Research Polytechnic University, Perm, Russia
| | - Galina Smirnova
- Laboratory of Physiology and Genetics of Microorganisms, Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Russian Academy of Sciences, Perm, Russia
| | - Olga Krasnykh
- Scientific and Educational Center for Applied Chemical and Biological Research, Perm National Research Polytechnic University, Perm, Russia
| | - Anastasija Boteva
- Scientific and Educational Center for Applied Chemical and Biological Research, Perm National Research Polytechnic University, Perm, Russia
| | - Oleg Oktyabrsky
- Laboratory of Physiology and Genetics of Microorganisms, Institute of Ecology and Genetics of Microorganisms, Perm Federal Research Center, Russian Academy of Sciences, Perm, Russia
| |
Collapse
|
3
|
Manikanta K, Paul M, Sandesha VD, Mahalingam SS, Ramesh TN, Harishkumar K, Koundinya SS, Naveen S, Kemparaju K, Girish KS. Oxidative Stress-Induced Platelet Apoptosis/Activation: Alleviation by Purified Curcumin via ASK1-JNK/p-38 Pathway. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:417-430. [PMID: 38648762 DOI: 10.1134/s0006297924030039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/26/2023] [Accepted: 01/29/2024] [Indexed: 04/25/2024]
Abstract
Platelets are known for their indispensable role in hemostasis and thrombosis. However, alteration in platelet function due to oxidative stress is known to mediate various health complications, including cardiovascular diseases and other health complications. To date, several synthetic molecules have displayed antiplatelet activity; however, their uses are associated with bleeding and other adverse effects. The commercially available curcumin is generally a mixture of three curcuminoids: curcumin, demethoxycurcumin, and bisdemethoxycurcumin. Although crude curcumin is known to inhibit platelet aggregation, the effect of purified curcumin on platelet apoptosis, activation, and aggregation remains unclear. Therefore, in this study, curcumin was purified from a crude curcumin mixture and the effects of this preparation on the oxidative stress-induced platelet apoptosis and activation was evaluated. 2,2'-Azobis(2-methylpropionamidine) dihydrochloride (AAPH) compound was used as an inducer of oxidative stress. Purified curcumin restored AAPH-induced platelet apoptotic markers like reactive oxygen species, intracellular calcium level, mitochondrial membrane potential, cardiolipin peroxidation, cytochrome c release from mitochondria to the cytosol, and phosphatidyl serine externalization. Further, it inhibited the agonist-induced platelet activation and aggregation, demonstrating its antiplatelet activity. Western blot analysis confirms protective effect of the purified curcumin against oxidative stress-induced platelet apoptosis and activation via downregulation of MAPKs protein activation, including ASK1, JNK, and p-38. Together, these results suggest that the purified curcumin could be a potential therapeutic bioactive molecule to treat the oxidative stress-induced platelet activation, apoptosis, and associated complications.
Collapse
Affiliation(s)
- Kurnegala Manikanta
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, 570006, India
| | - Manoj Paul
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, 570006, India
| | | | - Shanmuga S Mahalingam
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thimmasandra Narayan Ramesh
- Department of Studies and Research in Chemistry, University College of Science, Tumkur University, Tumakuru, 572103, India
| | | | - Shashank S Koundinya
- All India Institute of Medical Science, Sri Aurobindo Marg, Ansari Nagar, East, New Delhi, 110029, India
| | - Shivanna Naveen
- Applied Nutrition Discipline, Defense Food Research Laboratory, Mysuru, 570011, India
| | - Kempaiah Kemparaju
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, 570006, India.
| | - Kesturu S Girish
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru, 572103, India.
| |
Collapse
|
4
|
Babkina I, Savinkova I, Molchanova T, Sidorova M, Surin A, Gorbacheva L. Neuroprotective Effects of Noncanonical PAR1 Agonists on Cultured Neurons in Excitotoxicity. Int J Mol Sci 2024; 25:1221. [PMID: 38279219 PMCID: PMC10816171 DOI: 10.3390/ijms25021221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
Serine proteases regulate cell functions through G protein-coupled protease-activated receptors (PARs). Cleavage of one peptide bond of the receptor amino terminus results in the formation of a new N-terminus ("tethered ligand") that can specifically interact with the second extracellular loop of the PAR receptor and activate it. Activation of PAR1 by thrombin (canonical agonist) and activated protein C (APC, noncanonical agonist) was described as a biased agonism. Here, we have supposed that synthetic peptide analogs to the PAR1 tethered ligand liberated by APC could have neuroprotective effects like APC. To verify this hypothesis, a model of the ischemic brain impairment based on glutamate (Glu) excitotoxicity in primary neuronal cultures of neonatal rats has been used. It was shown that the nanopeptide NPNDKYEPF-NH2 (AP9) effectively reduced the neuronal death induced by Glu. The influence of AP9 on cell survival was comparable to that of APC. Both APC and AP9 reduced the dysregulation of intracellular calcium homeostasis in cultured neurons induced by excitotoxic Glu (100 µM) or NMDA (200 µM) concentrations. PAR1 agonist synthetic peptides might be noncanonical PAR1 agonists and a basis for novel neuroprotective drugs for disorders related to Glu excitotoxicity such as brain ischemia, trauma and some neurodegenerative diseases.
Collapse
Affiliation(s)
- Irina Babkina
- Faculty of Medical Biology, Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (I.B.); (I.S.)
| | - Irina Savinkova
- Faculty of Medical Biology, Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (I.B.); (I.S.)
| | - Tatiana Molchanova
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Maria Sidorova
- Chazov National Medical Research Center for Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia;
| | - Alexander Surin
- Laboratory of Fundamental and Applied Problems of Pain, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Liubov Gorbacheva
- Faculty of Medical Biology, Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia; (I.B.); (I.S.)
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| |
Collapse
|
5
|
Szabo E, Nagy B, Czajlik A, Komlodi T, Ozohanics O, Tretter L, Ambrus A. Mitochondrial Alpha-Keto Acid Dehydrogenase Complexes: Recent Developments on Structure and Function in Health and Disease. Subcell Biochem 2024; 104:295-381. [PMID: 38963492 DOI: 10.1007/978-3-031-58843-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The present work delves into the enigmatic world of mitochondrial alpha-keto acid dehydrogenase complexes discussing their metabolic significance, enzymatic operation, moonlighting activities, and pathological relevance with links to underlying structural features. This ubiquitous family of related but diverse multienzyme complexes is involved in carbohydrate metabolism (pyruvate dehydrogenase complex), the citric acid cycle (α-ketoglutarate dehydrogenase complex), and amino acid catabolism (branched-chain α-keto acid dehydrogenase complex, α-ketoadipate dehydrogenase complex); the complexes all function at strategic points and also participate in regulation in these metabolic pathways. These systems are among the largest multienzyme complexes with at times more than 100 protein chains and weights ranging up to ~10 million Daltons. Our chapter offers a wealth of up-to-date information on these multienzyme complexes for a comprehensive understanding of their significance in health and disease.
Collapse
Affiliation(s)
- Eszter Szabo
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Balint Nagy
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Andras Czajlik
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Timea Komlodi
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Oliver Ozohanics
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Laszlo Tretter
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Attila Ambrus
- Department of Biochemistry, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
6
|
Xu X, Xu S, Wan J, Wang D, Pang X, Gao Y, Ni N, Chen D, Sun X. Disturbing cytoskeleton by engineered nanomaterials for enhanced cancer therapeutics. Bioact Mater 2023; 29:50-71. [PMID: 37621771 PMCID: PMC10444958 DOI: 10.1016/j.bioactmat.2023.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 08/26/2023] Open
Abstract
Cytoskeleton plays a significant role in the shape change, migration, movement, adhesion, cytokinesis, and phagocytosis of tumor cells. In clinical practice, some anti-cancer drugs achieve cytoskeletal therapeutic effects by acting on different cytoskeletal protein components. However, in the absence of cell-specific targeting, unnecessary cytoskeletal recombination in organisms would be disastrous, which would also bring about severe side effects during anticancer process. Nanomedicine have been proven to be superior to some small molecule drugs in cancer treatment due to better stability and targeting, and lower side effects. Therefore, this review summarized the recent developments of various nanomaterials disturbing cytoskeleton for enhanced cancer therapeutics, including carbon, noble metals, metal oxides, black phosphorus, calcium, silicon, polymers, peptides, and metal-organic frameworks, etc. A comprehensive analysis of the characteristics of cytoskeleton therapy as well as the future prospects and challenges towards clinical application were also discussed. We aim to drive on this emerging topic through refreshing perspectives based on our own work and what we have also learnt from others. This review will help researchers quickly understand relevant cytoskeletal therapeutic information to further advance the development of cancer nanomedicine.
Collapse
Affiliation(s)
- Xueli Xu
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Shanbin Xu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Jipeng Wan
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Diqing Wang
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Xinlong Pang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yuan Gao
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore
| | - Dawei Chen
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xiao Sun
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| |
Collapse
|
7
|
Azhar NA, Abu Bakar SA, Ngalim SH, Ahmad NH. Cellular Uptake of Catharanthus roseus-Silver Nanoparticles in Human Hepatocellular Carcinoma HepG2 Cells. MALAYSIAN JOURNAL OF MEDICINE AND HEALTH SCIENCES 2023; 19:171-177. [DOI: 10.47836/mjmhs.19.4.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2024]
Abstract
Introduction: Nanoparticles exhibit unique features and currently at the forefront of cutting-edge research. Silver nanoparticles (AgNPs) are among the most promising and widely commercialised nanoproducts in various fields. The interaction of these AgNPs with cells remain unclear to connect with its toxicological endpoints. The aim of this study was to investigate the cellular uptake of C. roseus-AgNPs in hepatocellular carcinoma cells HepG2. Methods: The HepG2 cells were treated with the mean IC50 value of C. roseus-AgNPs which was 4.95±0.26 µg/mL for 24, 48 and 72 hours. The effects were compared with the untreated cells and other treatments which include camptothecin, C. roseus-aqueous extract, and AgNO3. Inductively coupled plasma optical emission spectroscopy (ICP-OES) was used to quantify the intracellular Ag+ and Ca2+, while transmission electron microscopy (TEM) imaging was used to visualise the nanoparticle distribution. Results: The HepG2 cells have significantly taken up Ag+ from C. roseus-AgNPs with at least six times higher compared to Ag+ from AgNO3. The intracellular Ca2+ detected in HepG2 cells for all treatments were significantly higher than the untreated cells, in time-dependent manner. TEM images indicated the endocytosis of C. roseus-AgNPs with the presence of endosomes and exocytic vesicles. Conclusion: The significant accumulation of intracellular Ag+ demonstrated the efficiency of the C. roseus-AgNPs uptake while the increased Ca2+ indicated the early sign of cell injury. The cellular uptake was mainly through endocytosis. These findings are crucial to correlate the physicochemical properties of C. roseus-AgNPs with the anticancer mechanisms towards the development of liver cancer therapy.
Collapse
|
8
|
Wang HN, Qian WJ, Zhao GL, Li F, Miao YY, Lei B, Sun XH, Wang ZF. L- and T-type Ca 2+ channels dichotomously contribute to retinal ganglion cell injury in experimental glaucoma. Neural Regen Res 2023; 18:1570-1577. [PMID: 36571364 PMCID: PMC10075096 DOI: 10.4103/1673-5374.360277] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Retinal ganglion cell apoptotic death is the main pathological characteristic of glaucoma, which is the leading cause of irreversible blindness. Disruption of Ca2+ homeostasis plays an important role in glaucoma. Voltage-gated Ca2+ channel blockers have been shown to improve vision in patients with glaucoma. However, whether and how voltage-gated Ca2+ channels are involved in retinal ganglion cell apoptotic death are largely unknown. In this study, we found that total Ca2+ current densities in retinal ganglion cells were reduced in a rat model of chronic ocular hypertension experimental glaucoma, as determined by whole-cell patch-clamp electrophysiological recordings. Further analysis showed that L-type Ca2+ currents were downregulated while T-type Ca2+ currents were upregulated at the later stage of glaucoma. Western blot assay and immunofluorescence experiments confirmed that expression of the CaV1.2 subunit of L-type Ca2+ channels was reduced and expression of the CaV3.3 subunit of T-type Ca2+ channels was increased in retinas of the chronic ocular hypertension model. Soluble tumor necrosis factor-α, an important inflammatory factor, inhibited the L-type Ca2+ current of isolated retinal ganglion cells from control rats and enhanced the T-type Ca2+ current. These changes were blocked by the tumor necrosis factor-α inhibitor XPro1595, indicating that both types of Ca2+ currents may be mediated by soluble tumor necrosis factor-α. The intracellular mitogen-activated protein kinase/extracellular signal-regulated kinase pathway and nuclear factor kappa-B signaling pathway mediate the effects of tumor necrosis factor-α. TUNEL assays revealed that mibefradil, a T-type calcium channel blocker, reduced the number of apoptotic retinal ganglion cells in the rat model of chronic ocular hypertension. These results suggest that T-type Ca2+ channels are involved in disrupted Ca2+ homeostasis and apoptosis of retinal ganglion cells in glaucoma, and application of T-type Ca2+ channel blockers, especially a specific CaV3.3 blocker, may be a potential strategy for the treatment of glaucoma.
Collapse
Affiliation(s)
- Hong-Ning Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wen-Jing Qian
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Guo-Li Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Fang Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yan-Ying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Bo Lei
- Institutes of Neuroscience and Third Affiliated Hospital, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xing-Huai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Zhong-Feng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Chen YM, Liu PY, Tang KT, Liu HJ, Liao TL. TWEAK-Fn14 Axis Induces Calcium-Associated Autophagy and Cell Death To Control Mycobacterial Survival in Macrophages. Microbiol Spectr 2022; 10:e0317222. [PMID: 36321903 PMCID: PMC9769850 DOI: 10.1128/spectrum.03172-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022] Open
Abstract
Autophagy is a natural defense mechanism that protects the host against pathogens. We previously demonstrated that mycobacterial infection upregulated tumor necrosis factor-like weak inducer of apoptosis (TWEAK) to promote autophagy and mycobacterial autophagosome maturation through activation of AMP-activated protein kinase (AMPK). Fibroblast growth factor-inducible 14 (Fn14) is the receptor of TWEAK. But the role of Fn14 in mycobacterial infection remains elusive. Herein, we observed increased expression of Fn14 in peripheral blood mononuclear cells of active tuberculosis (TB) patients. Downregulation of cellular Fn14 enhanced mycobacterial survival in macrophages. Conversely, Fn14 overexpression inhibited mycobacterial growth, suggesting that Fn14 can inhibit mycobacterial infection. The in vitro results revealed that TWEAK-promoted mycobacterial phagosome maturation is Fn14-dependent. We demonstrated that TWEAK-Fn14 signaling promotes oxidative stress to enhance the expression of stromal interaction molecule 1 (STIM1) and its activation of the Ca2+ channel ORAI1. Elevated calcium influx stimulated the activation of CaMCCK2 (calcium/calmodulin-dependent protein kinase kinase 2) and its downstream effector AMPK, thus inducing autophagy in early infection. Persistently TWEAK-Fn14 signaling caused cell death in late infection by reducing mitochondrial membrane potential, leading to mitochondrial ROS accumulation, and activating cell death-associated proteins. Genetic Fn14 deficiency or TWEAK blockers decreased oxidative stress-induced calcium influx, thus suppressing autophagy and cell death in mycobacteria-infected macrophages, and resulting in elevated mycobacterial survival. We propose that the TWEAK-Fn14 axis and calcium influx could be manipulated for anti-TB therapeutic purposes. Our results offer a new molecular machinery to understand the association between the TWEAK-Fn14 axis, calcium influx, and mycobacterial infection. IMPORTANCE Tuberculosis remains a major cause of morbidity and mortality worldwide. We previously demonstrated a relationship between TWEAK and activation of the autophagic machinery, which promotes anti-mycobacterial immunity. The TWEAK-Fn14 axis is multi-functional and involved in the pathogenesis of many diseases, thus blockade of TWEAK-Fn14 axis has been considered as a potential therapeutic target. Here, we demonstrated that the TWEAK-Fn14 axis plays a novel role in anti-mycobacterial infection by regulating calcium-associated autophagy. Persistently, TWEAK-Fn14 signaling caused cell death in late infection by reducing mitochondrial membrane potential, leading to mitochondrial ROS accumulation, and activating cell death-associated proteins. TWEAK blocker or Fn14 deficiency could suppress oxidative stress and calcium-associated autophagy, resulting in elevated mycobacterial survival. We propose that the TWEAK-Fn14 axis and calcium influx could be manipulated for anti-TB therapeutic purposes. This study offers a new molecular machinery to understand the association between the TWEAK-Fn14 axis, calcium influx, and mycobacterial infection.
Collapse
Affiliation(s)
- Yi-Ming Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Po-Yu Liu
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Division of Infection, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Kuo-Tung Tang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | - Hung-Jen Liu
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan, Republic of China
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Tsai-Ling Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
- Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan, Republic of China
| |
Collapse
|
10
|
Costa G, Ribeiro FF, Sebastião AM, Muir EM, Vaz SH. Bridging the gap of axonal regeneration in the central nervous system: A state of the art review on central axonal regeneration. Front Neurosci 2022; 16:1003145. [PMID: 36440273 PMCID: PMC9682039 DOI: 10.3389/fnins.2022.1003145] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/19/2022] [Indexed: 08/26/2023] Open
Abstract
Neuronal regeneration in the central nervous system (CNS) is an important field of research with relevance to all types of neuronal injuries, including neurodegenerative diseases. The glial scar is a result of the astrocyte response to CNS injury. It is made up of many components creating a complex environment in which astrocytes play various key roles. The glial scar is heterogeneous, diverse and its composition depends upon the injury type and location. The heterogeneity of the glial scar observed in different situations of CNS damage and the consequent implications for axon regeneration have not been reviewed in depth. The gap in this knowledge will be addressed in this review which will also focus on our current understanding of central axonal regeneration and the molecular mechanisms involved. The multifactorial context of CNS regeneration is discussed, and we review newly identified roles for components previously thought to solely play an inhibitory role in central regeneration: astrocytes and p75NTR and discuss their potential and relevance for deciding therapeutic interventions. The article ends with a comprehensive review of promising new therapeutic targets identified for axonal regeneration in CNS and a discussion of novel ways of looking at therapeutic interventions for several brain diseases and injuries.
Collapse
Affiliation(s)
- Gonçalo Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Filipa F. Ribeiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana M. Sebastião
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Elizabeth M. Muir
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Sandra H. Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
11
|
Basilico B, Palamà IE, D’Amone S, Lauro C, Rosito M, Grieco M, Ratano P, Cordella F, Sanchini C, Di Angelantonio S, Ragozzino D, Cascione M, Gigli G, Cortese B. Substrate stiffness effect on molecular crosstalk of epithelial-mesenchymal transition mediators of human glioblastoma cells. Front Oncol 2022; 12:983507. [PMID: 36091138 PMCID: PMC9454310 DOI: 10.3389/fonc.2022.983507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
The complexity of the microenvironment effects on cell response, show accumulating evidence that glioblastoma (GBM) migration and invasiveness are influenced by the mechanical rigidity of their surroundings. The epithelial–mesenchymal transition (EMT) is a well-recognized driving force of the invasive behavior of cancer. However, the primary mechanisms of EMT initiation and progression remain unclear. We have previously showed that certain substrate stiffness can selectively stimulate human GBM U251-MG and GL15 glioblastoma cell lines motility. The present study unifies several known EMT mediators to uncover the reason of the regulation and response to these stiffnesses. Our results revealed that changing the rigidity of the mechanical environment tuned the response of both cell lines through change in morphological features, epithelial-mesenchymal markers (E-, N-Cadherin), EGFR and ROS expressions in an interrelated manner. Specifically, a stiffer microenvironment induced a mesenchymal cell shape, a more fragmented morphology, higher intracellular cytosolic ROS expression and lower mitochondrial ROS. Finally, we observed that cells more motile showed a more depolarized mitochondrial membrane potential. Unravelling the process that regulates GBM cells’ infiltrative behavior could provide new opportunities for identification of new targets and less invasive approaches for treatment.
Collapse
Affiliation(s)
| | - Ilaria Elena Palamà
- National Research Council-Nanotechnology Institute (CNR Nanotec), Lecce, Italy
| | - Stefania D’Amone
- National Research Council-Nanotechnology Institute (CNR Nanotec), Lecce, Italy
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Maria Rosito
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
- Center for Life Nanoscience, Italian Institute of Technology (IIT), Rome, Italy
| | - Maddalena Grieco
- National Research Council-Nanotechnology Institute (CNR Nanotec), Lecce, Italy
| | - Patrizia Ratano
- National Research Council-Nanotechnology Institute (CNR Nanotec), Rome, Italy
| | - Federica Cordella
- Center for Life Nanoscience, Italian Institute of Technology (IIT), Rome, Italy
| | - Caterina Sanchini
- Center for Life Nanoscience, Italian Institute of Technology (IIT), Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
- Center for Life Nanoscience, Italian Institute of Technology (IIT), Rome, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | - Giuseppe Gigli
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
- Department of Mathematics and Physics “Ennio De Giorgi” University of Salento, Lecce, Italy
| | - Barbara Cortese
- National Research Council-Nanotechnology Institute (CNR Nanotec), Rome, Italy
- *Correspondence: Barbara Cortese,
| |
Collapse
|
12
|
Mohammadnejad L, Soltaninejad K, Seyedabadi M, Ghasem Pouri SK, Shokrzadeh M, Mohammadi H. Evaluation of mitochondrial dysfunction due to oxidative stress in therapeutic, toxic and lethal concentrations of tramadol. Toxicol Res (Camb) 2021; 10:1162-1170. [PMID: 34956619 DOI: 10.1093/toxres/tfab096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/01/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Tramadol (TR) is a centrally acting analgesic drug that is used to relieve pain. The therapeutic (0.1-0.8 mg/l), toxic (1-2 mg/l) and lethal (>2 mg/l) ranges were reported for TR. The present study was designed to evaluate which doses of TR can induce liver mitochondrial toxicity. Mitochondria were isolated from the five rats' liver and were incubated with therapeutic to lethal concentrations (1.7-600 μM) of TR. Biomarkers of oxidative stress including: reactive oxygen species (ROS), lipid peroxidation (LPO), protein carbonyl content, glutathione (GSH) content, mitochondrial function, mitochondrial membrane potential (MMP) and mitochondrial swelling were assessed. Our results showed that ROS and LPO at 100 μM and protein carbonylation at 600 μM concentrations of TR were significantly increased. GSH was decreased specifically at 600 μM concentration. Mitochondrial function, MMP and mitochondrial swelling decreased in isolated rat liver mitochondria after exposure to 100 and 300 μM, respectively. This study suggested that TR at therapeutic and toxic levels by single exposure could not induce mitochondrial toxicity. But, in lethal concentration (≥100 μM), TR induced oxidative damage and mitochondria dysfunction. This study suggested that ROS overproduction by increasing of TR concentration induced mitochondrial dysfunction and caused mitochondrial damage via Complex II and membrane permeability transition pores disorders, MMP collapse and mitochondria swelling.
Collapse
Affiliation(s)
- Leila Mohammadnejad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Kambiz Soltaninejad
- Department of Forensic Toxicology, Legal Medicine Research Center, Legal Medicine Organization, Tehran 48157-33971, Iran
| | - Mohammad Seyedabadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Seyed Khosro Ghasem Pouri
- Department of Emergency Medicine, School of Medicine, Antimicrobial Resistance Research Center, Ghaem Shahr Razi Hospital, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Mohammad Shokrzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Hamidreza Mohammadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| |
Collapse
|
13
|
Goshtasbi H, Pakchin PS, Movafeghi A, Barar J, Castejon AM, Omidian H, Omidi Y. Impacts of oxidants and antioxidants on the emergence and progression of Alzheimer's disease. Neurochem Int 2021; 153:105268. [PMID: 34954260 DOI: 10.1016/j.neuint.2021.105268] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/29/2021] [Accepted: 12/21/2021] [Indexed: 01/06/2023]
Abstract
The brain shows a high sensitivity to oxidative stress (OS). Thus, the maintenance of homeostasis of the brain regarding the reduction-oxidation (redox) situation is crucial for the regular function of the central nervous systems (CNS). The imbalance between the reactive oxygen species (ROS) and the cellular mechanism might lead to the emergence of OS, causing profound cell death as well as tissue damages and initiating neurodegenerative disorders (NDDs). Characterized by the cytoplasmic growth of neurofibrillary tangles and extracellular β-amyloid plaques, Alzheimer's disease (AD) is a complex NDD that causes dementia in adult life with severe manifestations. Nuclear factor erythroid 2-related factor 2 (NRF2) is a key transcription factor that regulates the functional expression of OS-related genes and the functionality of endogenous antioxidants. In the case of oxidative damage, NRF2 is transferred to the nucleus and attached to the antioxidant response element (ARE) that enhances the sequence to initiate transcription of the cell-protecting genes. This review articulates various mechanisms engaged with the generation of active and reactive species of endogenous and exogenous oxidants and focuses on the antioxidants as a body defense system regarding the NRF2-ARE signaling path in the CNS.
Collapse
Affiliation(s)
- Hamieh Goshtasbi
- Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Samadi Pakchin
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Movafeghi
- Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ana M Castejon
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, United States
| | - Hossein Omidian
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, United States
| | - Yadollah Omidi
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, United States.
| |
Collapse
|
14
|
Ergan Sahin A, Karasoy Yesilada A, Yalcin O, Guler EM, Erbek H, Karabıyık D. Hydrogen-rich saline reduces tissue injury and improves skin flap survival on a rat hindlimb degloving injury model. J Plast Reconstr Aesthet Surg 2021; 74:2095-2103. [PMID: 33451944 DOI: 10.1016/j.bjps.2020.12.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/01/2020] [Accepted: 12/17/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Degloving injuries represent a challenge in plastic surgery. The aim of this study is to acknowledge the protective effects of hydrogen-rich saline (HRS) solution on a rat hindlimb degloved skin flap. METHODS Twenty-one Sprague-Dawley rats were divided into three groups (control, saline and HRS). Degloving injury model was established, and flaps were sutured back following 5 min of ischemia. The control group did not receive any treatment. The saline group received intraperitoneal physiological saline (10 ml/kg) and the HRS group received intraperitoneal HRS solution (10 ml/kg) postoperatively and daily for 5 days after the operation. Skin samples were obtained for histological, immunohistochemical and biochemical evaluations. RESULTS Inflammation was lower in the HRS compared with saline (p = 0.02) and control (p = 0.004) groups. Edema was lower in the HRS compared with saline (p = 0.02) and control (p = 0.001) groups. Malondialdehyde (MDA) level was lower in the HRS than the control group (p = 0.01). Total antioxidant level was higher in the HRS compared with saline (p = 0.009) and control (p = 0.03) groups. Total oxidant level was lower in the HRS than the control group (p = 0.02). Oxidative stress index was lower in the HRS compared with saline (p = 0.001) and control (p = 0.0001) groups`. Vascular proliferation was higher in the HRS compared with the control group (p = 0.01). CONCLUSION Repeated HRS injections after trauma increased the viability of skin flap in rat degloving injury model by decreasing local tissue injury, due to its antioxidant, anti-inflammatory and angiogenic effects.
Collapse
Affiliation(s)
- Ayca Ergan Sahin
- Department of Plastic Surgery, Prof. Dr. Cemil Tascioglu City Hospital, University of Health Sciences, Istanbul, Turkey.
| | - Aysin Karasoy Yesilada
- Department of Plastic Surgery, Medipol Healthcare Group, Camlica Medipol University Hospital, Istanbul, Turkey
| | - Ozben Yalcin
- Department of Pathology, Prof. Dr. Cemil Tascioglu City Hospital, University of Health Sciences, Istanbul, Turkey
| | - Eray M Guler
- Health Sciences University Hamidiye Medicine Faculty Department of Medical Biochemistry, Istanbul, Turkey
| | - Harun Erbek
- Department of Plastic Surgery, Prof. Dr. Cemil Tascioglu City Hospital, University of Health Sciences, Istanbul, Turkey
| | - Damla Karabıyık
- Department of Pathology, Prof. Dr. Cemil Tascioglu City Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
15
|
Bessone F, Hernández N, Tanno M, Roma MG. Drug-Induced Vanishing Bile Duct Syndrome: From Pathogenesis to Diagnosis and Therapeutics. Semin Liver Dis 2021; 41:331-348. [PMID: 34130334 DOI: 10.1055/s-0041-1729972] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The most concerned issue in the context of drug/herb-induced chronic cholestasis is vanishing bile duct syndrome. The progressive destruction of intrahepatic bile ducts leading to ductopenia is usually not dose dependent, and has a delayed onset that should be suspected when abnormal serum cholestasis enzyme levels persist despite drug withdrawal. Immune-mediated cholangiocyte injury, direct cholangiocyte damage by drugs or their metabolites once in bile, and sustained exposure to toxic bile salts when biliary epithelium protective defenses are impaired are the main mechanisms of cholangiolar damage. Current therapeutic alternatives are scarce and have not shown consistent beneficial effects so far. This review will summarize the current literature on the main diagnostic tools of ductopenia and its histological features, and the differential diagnostic with other ductopenic diseases. In addition, pathomechanisms will be addressed, as well as the connection between them and the supportive and curative strategies for ductopenia management.
Collapse
Affiliation(s)
- Fernando Bessone
- Hospital Provincial del Centenario, Facultad de Ciencias Médicas, Servicio de Gastroenterología y Hepatología, Universidad Nacional de Rosario, Rosario, Argentina
| | - Nelia Hernández
- Clínica de Gastroenterología, Hospital de Clínicas y Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Mario Tanno
- Hospital Provincial del Centenario, Facultad de Ciencias Médicas, Servicio de Gastroenterología y Hepatología, Universidad Nacional de Rosario, Rosario, Argentina
| | - Marcelo G Roma
- Instituto de Fisiología Experimental (CONICET-UNR), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
16
|
Wang H, Zhang Y, Xin F, Jiang H, Tao D, Jin Y, He Y, Wang Q, Po SS. Calcium-Induced Autonomic Denervation in Patients With Post-Operative Atrial Fibrillation. J Am Coll Cardiol 2021; 77:57-67. [PMID: 33413942 DOI: 10.1016/j.jacc.2020.10.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 10/14/2020] [Accepted: 11/10/2020] [Indexed: 01/16/2023]
Abstract
BACKGROUND Post-operative atrial fibrillation (POAF) is associated with worse long-term cardiovascular outcomes. OBJECTIVES This study hypothesized that injecting calcium chloride (CaCl2) into the major atrial ganglionated plexi (GPs) during isolated coronary artery bypass grafting (CABG) can reduce the incidence of POAF by calcium-induced autonomic neurotoxicity. METHODS This proof-of-concept study randomized 200 patients undergoing isolated, off-pump CABG to CaCl2 (n = 100) or sodium chloride (sham, n = 100) injection. Two milliliters of CaCl2 (5%) or sodium chloride (0.9%) was injected into the 4 major atrial GPs during CABG. All patients received 7-day continuous telemetry and Holter monitoring. The primary outcome was incidence of POAF (≥30 s) in 7 days. Secondary outcomes included length of hospitalization, POAF burden, average ventricular rate during AF, plasma level of inflammatory markers, and actionable antiarrhythmic therapy to treat POAF. RESULTS The POAF incidence was reduced from 36% to 15% (hazard ratio: 0.366; 95% confidence interval: 0.211 to 0.635; p = 0.001). Length of hospitalization did not differ between the 2 groups. POAF burden (first 7 post-operative days), the use of amiodarone or esmolol, and the incidence of atrial couplets and nonsustained atrial tachyarrhythmias were significantly reduced in the CaCl2 group. Heart rate variability data showed a decrease in both high-frequency and low-frequency power in the CaCl2 group with a preserved low-frequency/high-frequency ratio, suggesting that the sympathetic/parasympathetic balance was not perturbed by CaCl2 injection. CONCLUSIONS Injection of CaCl2 into the 4 major atrial GPs reduced the POAF hazard by 63%. Inhibition of GP function by Ca-mediated neurotoxicity may underlie the therapeutic effect. (Calcium Autonomic Denervation Prevents Postoperative Atrial Fibrillation; ChiCTR1800019276).
Collapse
Affiliation(s)
- Huishan Wang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China.
| | - Yuji Zhang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Fangran Xin
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Hui Jiang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Dengshun Tao
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Yan Jin
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Yuanchen He
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Qiang Wang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Sunny S Po
- Section of Cardiovascular Diseases and Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.
| |
Collapse
|
17
|
Sciaccaluga M, Megaro A, Bellomo G, Ruffolo G, Romoli M, Palma E, Costa C. An Unbalanced Synaptic Transmission: Cause or Consequence of the Amyloid Oligomers Neurotoxicity? Int J Mol Sci 2021; 22:ijms22115991. [PMID: 34206089 PMCID: PMC8199544 DOI: 10.3390/ijms22115991] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022] Open
Abstract
Amyloid-β (Aβ) 1-40 and 1-42 peptides are key mediators of synaptic and cognitive dysfunction in Alzheimer's disease (AD). Whereas in AD, Aβ is found to act as a pro-epileptogenic factor even before plaque formation, amyloid pathology has been detected among patients with epilepsy with increased risk of developing AD. Among Aβ aggregated species, soluble oligomers are suggested to be responsible for most of Aβ's toxic effects. Aβ oligomers exert extracellular and intracellular toxicity through different mechanisms, including interaction with membrane receptors and the formation of ion-permeable channels in cellular membranes. These damages, linked to an unbalance between excitatory and inhibitory neurotransmission, often result in neuronal hyperexcitability and neural circuit dysfunction, which in turn increase Aβ deposition and facilitate neurodegeneration, resulting in an Aβ-driven vicious loop. In this review, we summarize the most representative literature on the effects that oligomeric Aβ induces on synaptic dysfunction and network disorganization.
Collapse
Affiliation(s)
- Miriam Sciaccaluga
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
- Correspondence: (M.S.); (C.C.); Tel.: +39-0755858180 (M.S.); +39-0755784233 (C.C.)
| | - Alfredo Megaro
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
| | - Giovanni Bellomo
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy; (G.R.); (E.P.)
- IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Michele Romoli
- Neurology Unit, Rimini “Infermi” Hospital—AUSL Romagna, 47923 Rimini, Italy;
| | - Eleonora Palma
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy; (G.R.); (E.P.)
| | - Cinzia Costa
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
- Correspondence: (M.S.); (C.C.); Tel.: +39-0755858180 (M.S.); +39-0755784233 (C.C.)
| |
Collapse
|
18
|
Photobiomodulation and Oxidative Stress: 980 nm Diode Laser Light Regulates Mitochondrial Activity and Reactive Oxygen Species Production. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6626286. [PMID: 33763170 PMCID: PMC7952159 DOI: 10.1155/2021/6626286] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/09/2021] [Accepted: 02/17/2021] [Indexed: 12/20/2022]
Abstract
Photobiomodulation with 808 nm laser light electively stimulates Complexes III and IV of the mitochondrial respiratory chain, while Complexes I and II are not affected. At the wavelength of 1064 nm, Complexes I, III, and IV are excited, while Complex II and some mitochondrial matrix enzymes seem to be not receptive to photons at that wavelength. Complex IV was also activated by 633 nm. The mechanism of action of wavelengths in the range 900–1000 nm on mitochondria is less understood or not described. Oxidative stress from reactive oxygen species (ROS) generated by mitochondrial activity is an inescapable consequence of aerobic metabolism. The antioxidant enzyme system for ROS scavenging can keep them under control. However, alterations in mitochondrial activity can cause an increment of ROS production. ROS and ATP can play a role in cell death, cell proliferation, and cell cycle arrest. In our work, bovine liver isolated mitochondria were irradiated for 60 sec, in continuous wave mode with 980 nm and powers from 0.1 to 1.4 W (0.1 W increment at every step) to generate energies from 6 to 84 J, fluences from 7.7 to 107.7 J/cm2, power densities from 0.13 to 1.79 W/cm2, and spot size 0.78 cm2. The control was equal to 0 W. The activity of the mitochondria's complexes, Krebs cycle enzymes, ATP production, oxygen consumption, generation of ROS, and oxidative stress were detected. Lower powers (0.1–0.2 W) showed an inhibitory effect; those that were intermediate (0.3–0.7 W) did not display an effect, and the higher powers (0.8–1.1 W) induced an increment of ATP synthesis. Increasing the power (1.2–1.4 W) recovered the ATP production to the control level. The interaction occurred on Complexes III and IV, as well as ATP production and oxygen consumption. Results showed that 0.1 W uncoupled the respiratory chain and induced higher oxidative stress and drastic inhibition of ATP production. Conversely, 0.8 W kept mitochondria coupled and induced an increase of ATP production by increments of Complex III and IV activities. An augmentation of oxidative stress was also observed, probably as a consequence of the increased oxygen consumption and mitochondrial isolation experimental conditions. No effect was observed using 0.5 W, and no effect was observed on the enzymes of the Krebs cycle.
Collapse
|
19
|
Shen B, Wang S, Bharathi G, Li Y, Lin F, Hu R, Liu L, Qu J. Rapid and Targeted Photoactivation of Ca 2+ Channels Mediated by Squaraine To Regulate Intracellular and Intercellular Signaling Processes. Anal Chem 2020; 92:8497-8505. [PMID: 32438796 DOI: 10.1021/acs.analchem.0c01243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As an important cellular signal transduction messenger, Ca2+ has the capability to regulate cell function and control many biochemical processes, including metabolism, gene expression, and cell survival and death. Here, we introduce an accessible method for the photoactivation of Ca2+ channels mediated by squaraine (SQ) to rapidly induce cellular Ca2+ release and activate signal transduction. With a short preparation time, the maximum Ca2+ concentration increase could reach approximately 450% in 30 s, resulting from marked Ca2+ release channel opening in the endoplasmic reticulum (ER). This release was enhanced by another target location of SQ, that is, the outer mitochondrial-associated membrane where Ca2+ channels accumulate, and by the consequent large amounts of reactive oxygen species resulting from the respiratory chain activity stimulated by Ca2+ load. We used this method to investigate cellular signal transduction in different cancer cells and revealed rapid intracellular Ca2+ flow, unidirectional intercellular signaling processes, and neuronal signaling activity, which demonstrated the potential and convenience of the method for routine Ca2+ research.
Collapse
Affiliation(s)
- Binglin Shen
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Ganapathi Bharathi
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Yanping Li
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Fangrui Lin
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Rui Hu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| |
Collapse
|
20
|
Ponne S, Kumar CR, Boopathy R. Verapamil attenuates scopolamine induced cognitive deficits by averting oxidative stress and mitochondrial injury - A potential therapeutic agent for Alzheimer's Disease. Metab Brain Dis 2020; 35:503-515. [PMID: 31691145 DOI: 10.1007/s11011-019-00498-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 09/20/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial disorder where amyloid beta (Aβ) plaques, Ca2+ dysregulation, excessive oxidative stress, mitochondrial dysfunction and synaptic loss operate synergistically to bring about cholinergic deficits and dementia. New therapeutic interventions are gaining prominence as the morbidity and mortality of AD increases exponentially every year. Treating AD with antihypertensive drugs is thought to be a promising intervention; however, its mechanism of action of ameliorating AD needs further investigation. In this context, the present study explores the protective effect of verapamil, an antihypertensive agent of Ca2+ channel blocker (CCB) class against scopolamine-induced in vitro neurotoxicity and in vivo cognitive impairment. Supplementation of verapamil was found to attenuate oxidative stress by preventing mitochondrial injury, and augment the expression of genes involved in the cholinergic function (mACR1), synaptic plasticity (GAP43, SYP) and Ca2+-dependent memory-related genes (CREB1, CREBBP, BDNF). Further, verapamil treatment in mice attenuated the cognitive and behavioural deficits induced by scopolamine as measured by the elevated plus maze and passive avoidance test (P < 0.05). Thus, the present study demonstrates the neuroprotective effect of verapamil against the pathogenesis of AD such as oxidative stress, mitochondrial dysfunction and cognitive decline. These observations emphasize the importance of ‛Ca2+ dysregulation' and ‛mitochondrial dysfunction' theories in AD and recommends the supplementation of compounds that regulate Ca2+ homeostasis and mitochondrial function in susceptible AD individuals.
Collapse
Affiliation(s)
- Saravanaraman Ponne
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India.
- Department of Biotechnology, Pondicherry University, Puducherry, Kalapet, 605014, India.
| | - Chinnadurai Raj Kumar
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Rathanam Boopathy
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| |
Collapse
|
21
|
Zhu J, Ye Q, Xu S, Chang YX, Liu X, Ma Y, Zhu Y, Hua S. Shengmai injection alleviates H 2O 2‑induced oxidative stress through activation of AKT and inhibition of ERK pathways in neonatal rat cardiomyocytes. JOURNAL OF ETHNOPHARMACOLOGY 2019; 239:111677. [PMID: 30615921 DOI: 10.1016/j.jep.2019.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 12/07/2018] [Accepted: 01/02/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shengmai injection (SMI) is a classical traditional Chinese medicine (TCM) officially recorded in Pharmacopoeia of the People's Republic of China (version 2015) and has long been used to treat heart failure in China. However scientific evidence for the anti-oxidative stress potential of SMI used in traditional medicine is lacking. AIM OF STUDY The present study aimed to evaluate the efficacy of SMI in alleviating H2O2‑induced Oxidative Stress the underlying mechanisms MATERIALS AND METHODS: H2O2-induced oxidative stress model of cardiomyocytes was established with primary cultured neonatal rat cardiomyocytes. CCK8 cell viability assay and lacatate dehydrogenase cytotoxicity assay were performed to ensure the safety dose and lowest effective dose for the mode employing CCK-8 cell viability assay kit and lactate dehydrogenase cytotoxicity assay kit. ROS levels were determined using CM-H2DCFDA fluorescent probe in cardiomyocytes with H2O2-induced oxidative stress. The change of NAD(P)H level in cardiomyocytes was evaluated during the process of oxidative stress. The content of myocardial cytosolic Ca2+ and Ca2+ was determined using Fura-2/AM and Rhod 2-AM fluorescent probe in mitochondrial in the process of oxidative stress. Annexin V-FITC/PI double staining was applied to examine the apoptotic cells in cardiomyocytes with oxidative stress. To identify the apoptosis after oxidative stress myocardial cells with the application of Annexin V-FITC/PI double staining apoptosis detection kit. Quantitative polymerase chain reaction (RT-PCR) was applied to measure the expression of antioxidant enzymes: catalase (CAT), superoxide dismutase (SOD), glutathione reductase (GSR). Western blot was performed to observe the phosphorylation of AKT and ERK1/2. RESULTS SMI was shown to significantly attenuate oxidative stress-induced cell proliferation arrest and apoptosis in neonatal rat cardiomyocytes. In addition, SMI treatment could decrease the production of reactive oxygen species (ROS), nicotinamide adenine dinucleotide (NADH) and malondialdehyde (MDA), and reduce the overloads of cytoplasmic Ca2+ and mitochondrial Ca2+ induced by H2O2. SMI could also restore the mRNA expression and activities of SOD, GSR, and CAT suppressed by H2O2. Mechanistically, SMI upregulated intracellular AKT phosphorylation and downregulate ERK1/2 phosphorylation in H2O2-treated cardiomyocytes. Pretreatment with LY294002, an AKT phosphorylation inhibitor, suppressed the protective role of SMI in cardiomyocytes, while pretreatment with PD98059, an ERK1/2 phosphorylation inhibitor, enhanced the effect of SMI. CONCLUSIONS In conclusion, SMI may attenuate oxidative stress-induced damage in cardiomyocytes potentially through the AKT and ERK1/2 pathway and can function as a promising injectable traditional Chinese medicine to treat oxidative stress-induced injury.
Collapse
Affiliation(s)
- Jinqiang Zhu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Qiaofeng Ye
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 An Shan Xi Road, Nan Kai District, Tianjin 300193, China
| | - Yan-Xu Chang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuan Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yan Ma
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300150, China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin 300457, China
| | - Shengyu Hua
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; College of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
22
|
Zhou H, Niu L, Meng L, Lin Z, Zou J, Xia X, Huang X, Zhou W, Bian T, Zheng H. Noninvasive Ultrasound Deep Brain Stimulation for the Treatment of Parkinson's Disease Model Mouse. RESEARCH 2019; 2019:1748489. [PMID: 31549045 PMCID: PMC6750068 DOI: 10.34133/2019/1748489] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/22/2019] [Indexed: 01/22/2023]
Abstract
Modulating basal ganglia circuitry is of great significance in the improvement of motor function in Parkinson's disease (PD). Here, for the first time, we demonstrate that noninvasive ultrasound deep brain stimulation (UDBS) of the subthalamic nucleus (STN) or the globus pallidus (GP) improves motor behavior in a subacute mouse model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Immunohistochemical c-Fos protein expression confirms that there is a relatively high level of c-Fos expression in the STN-UDBS and GP-UDBS group compared with sham group (both p < 0.05). Furthermore, STN-UDBS or GP-UDBS significantly increases the latency to fall in the rotarod test on day 9 (p < 0.05) and decreases the time spent climbing down a vertical rod in the pole test on day 12 (p < 0.05). Moreover, our results reveal that STN-UDBS or GP-UDBS protects the dopamine (DA) neurons from MPTP neurotoxicity by downregulating Bax (p < 0.001), upregulating Bcl-2 (p < 0.01), blocking cytochrome c (Cyt C) release from mitochondria (p < 0.05), and reducing cleaved-caspase 3 activity (p < 0.01) in the ipsilateral substantia nigra (SN). Additionally, the safety of ultrasound stimulation is characterized by hematoxylin and eosin (HE) and Nissl staining; no hemorrhage or tissue damage is detected. These data demonstrate that UDBS enables modulation of STN or GP neural activity and leads to neuroprotection in PD mice, potentially serving as a noninvasive strategy for the clinical treatment of PD.
Collapse
Affiliation(s)
- Hui Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Long Meng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Zhengrong Lin
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Junjie Zou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Xiangxiang Xia
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Xiaowei Huang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Wei Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, China
| | - Tianyuan Bian
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| |
Collapse
|
23
|
Yang L, Dong Y, Wu C, Li Y, Guo Y, Yang B, Zong X, Hamblin MR, Cheng-Yi Liu T, Zhang Q. Photobiomodulation preconditioning prevents cognitive impairment in a neonatal rat model of hypoxia-ischemia. JOURNAL OF BIOPHOTONICS 2019; 12:e201800359. [PMID: 30652418 PMCID: PMC6546525 DOI: 10.1002/jbio.201800359] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/02/2018] [Accepted: 01/12/2019] [Indexed: 05/13/2023]
Abstract
Neonatal hypoxia-ischemia (HI) injury caused by oxygen deprivation is the most common cause of mortality and severe neurologic deficits in neonates. The present work evaluated the preventative effect of photobiomodulation (PBM) preconditioning, and its underlying mechanism of action on brain damage in an HI model in neonatal rats. According to the optimal time response of ATP levels in brain samples removed from normal rats, a PBM preconditioning (PBM-P) regimen (808 nm CW laser, 1 cm2 spot, 100 mW/cm2 , 12 J/cm2 ) was delivered to the scalp 6 hours before HI. PBM-P significantly attenuated cognitive impairment, volume shrinkage in the brain, neuron loss, dendritic and synaptic injury after HI. Further mechanistic investigation found that PBM-P could restore HI-induced mitochondrial dynamics and inhibit mitochondrial fragmentation, followed by a robust suppression of cytochrome c release, and prevention of neuronal apoptosis by inhibition of caspase activation. Our work suggests that PBM-P can attenuate HI-induced brain injury by maintaining mitochondrial dynamics and inhibiting the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Luodan Yang
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University, University Town, Guangzhou, GD 510006, China
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Yan Dong
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Chongyun Wu
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University, University Town, Guangzhou, GD 510006, China
| | - Yong Li
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Yichen Guo
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Baocheng Yang
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Xuemei Zong
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University, University Town, Guangzhou, GD 510006, China
| | - Quanguang Zhang
- Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University, University Town, Guangzhou, GD 510006, China
| |
Collapse
|
24
|
Ando S, Funato M, Ohuchi K, Inagaki S, Sato A, Seki J, Kawase C, Saito T, Nishio H, Nakamura S, Shimazawa M, Kaneko H, Hara H. The Protective Effects of Levetiracetam on a Human iPSCs-Derived Spinal Muscular Atrophy Model. Neurochem Res 2019; 44:1773-1779. [PMID: 31102025 DOI: 10.1007/s11064-019-02814-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/04/2019] [Accepted: 05/09/2019] [Indexed: 12/30/2022]
Abstract
Spinal muscular atrophy (SMA) is an inherited disease characterized by progressive motor neuron death and subsequent muscle weakness and is caused by deletion or mutation of survival motor neuron (SMN) 1 gene. Protecting spinal motor neuron is an effective clinical strategy for SMA. The purpose of this study was to investigate the potential effect of an anti-epileptic drug levetiracetam on SMA. In the present study, we used differentiated spinal motor neurons (MNs) from SMA patient-derived induced pluripotent stem cells (SMA-iPSCs) to investigate the effect of levetiracetam. Levetiracetam promoted neurite elongation in SMA-iPSCs-MNs. TUNEL-positive spinal motor neurons were significantly reduced by levetiracetam in SMA-iPSCs-MNs. In addition, the expression level of cleaved-caspase 3 was decreased by levetiracetam in SMA-iPSCs-MNs. Furthermore, levetiracetam improved impaired mitochondrial function in SMA-iPSCs-MNs. On the other hand, levetiracetam did not affect the expression level of SMN protein in SMA-iPSCs-MNs. These findings indicate that levetiracetam has a neuroprotective effect for SMA.
Collapse
Affiliation(s)
- Shiori Ando
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Michinori Funato
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Kazuki Ohuchi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Satoshi Inagaki
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Arisu Sato
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Junko Seki
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Chizuru Kawase
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Toshio Saito
- Division of Child Neurology, Department of Neurology, National Hospital Organization, Toneyama National Hospital, Toyonaka, Osaka, Japan
| | - Hisahide Nishio
- Department of Occupational Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe, Hyogo, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Hideo Kaneko
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan.
| |
Collapse
|
25
|
The Effects of Sodium Dichloroacetate on Mitochondrial Dysfunction and Neuronal Death Following Hypoglycemia-Induced Injury. Cells 2019; 8:cells8050405. [PMID: 31052436 PMCID: PMC6562710 DOI: 10.3390/cells8050405] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/17/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022] Open
Abstract
Our previous studies demonstrated that some degree of neuronal death is caused by hypoglycemia, but a subsequent and more severe wave of neuronal cell death occurs due to glucose reperfusion, which results from the rapid restoration of low blood glucose levels. Mitochondrial dysfunction caused by hypoglycemia leads to increased levels of pyruvate dehydrogenase kinase (PDK) and suppresses the formation of ATP by inhibiting pyruvate dehydrogenase (PDH) activation, which can convert pyruvate into acetyl-coenzyme A (acetyl-CoA). Sodium dichloroacetate (DCA) is a PDK inhibitor and activates PDH, the gatekeeper of glucose oxidation. However, no studies about the effect of DCA on hypoglycemia have been published. In the present study, we hypothesized that DCA treatment could reduce neuronal death through improvement of glycolysis and prevention of reactive oxygen species production after hypoglycemia. To test this, we used an animal model of insulin-induced hypoglycemia and injected DCA (100 mg/kg, i.v., two days) following hypoglycemic insult. Histological evaluation was performed one week after hypoglycemia. DCA treatment reduced hypoglycemia-induced oxidative stress, microglial activation, blood–brain barrier disruption, and neuronal death compared to the vehicle-treated hypoglycemia group. Therefore, our findings suggest that DCA may have the therapeutic potential to reduce hippocampal neuronal death after hypoglycemia.
Collapse
|
26
|
Cole AR, Perry DA, Raza A, Nedder AP, Pollack E, Regan WL, van den Bosch SJ, Polizzotti BD, Yang E, Davila D, Afacan O, Warfield SK, Ou Y, Sefton B, Everett AD, Neil JJ, Lidov HG, Mayer JE, Kheir JN. Perioperatively Inhaled Hydrogen Gas Diminishes Neurologic Injury Following Experimental Circulatory Arrest in Swine. JACC Basic Transl Sci 2019; 4:176-187. [PMID: 31061920 PMCID: PMC6488769 DOI: 10.1016/j.jacbts.2018.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 12/30/2022]
Abstract
This study used a swine model of mildly hypothermic prolonged circulatory arrest and found that the addition of 2.4% inhaled hydrogen gas to inspiratory gases during and after the ischemic insult significantly decreased neurologic and renal injury compared with controls. With proper precautions, inhalational hydrogen may be administered safely through conventional ventilators and may represent a complementary therapy that can be easily incorporated into current workflows. In the future, inhaled hydrogen may diminish the sequelae of ischemia that occurs in congenital heart surgery, cardiac arrest, extracorporeal life-support events, acute myocardial infarction, stroke, and organ transplantation.
Collapse
Affiliation(s)
- Alexis R. Cole
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
| | - Dorothy A. Perry
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Ali Raza
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Arthur P. Nedder
- Animal Resources at Children’s Hospital, Boston Children’s Hospital, Boston, Massachusetts
| | - Elizabeth Pollack
- Animal Resources at Children’s Hospital, Boston Children’s Hospital, Boston, Massachusetts
| | - William L. Regan
- Department of Cardiovascular Surgery, Boston Children’s Hospital, Boston, Massachusetts
| | | | - Brian D. Polizzotti
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Edward Yang
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Daniel Davila
- Department of Neurology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Onur Afacan
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Simon K. Warfield
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Yangming Ou
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Brenda Sefton
- Department of Cardiovascular Surgery, Boston Children’s Hospital, Boston, Massachusetts
| | - Allen D. Everett
- Division of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jeffrey J. Neil
- Department of Radiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Radiology, Harvard Medical School, Boston, Massachusetts
| | - Hart G.W. Lidov
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - John E. Mayer
- Department of Cardiovascular Surgery, Boston Children’s Hospital, Boston, Massachusetts
- Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - John N. Kheir
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
27
|
Núñez MT, Hidalgo C. Noxious Iron-Calcium Connections in Neurodegeneration. Front Neurosci 2019; 13:48. [PMID: 30809110 PMCID: PMC6379295 DOI: 10.3389/fnins.2019.00048] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/18/2019] [Indexed: 12/26/2022] Open
Abstract
Iron and calcium share the common feature of being essential for normal neuronal function. Iron is required for mitochondrial function, synaptic plasticity, and the development of cognitive functions whereas cellular calcium signals mediate neurotransmitter exocytosis, axonal growth and synaptic plasticity, and control the expression of genes involved in learning and memory processes. Recent studies have revealed that cellular iron stimulates calcium signaling, leading to downstream activation of kinase cascades engaged in synaptic plasticity. The relationship between calcium and iron is Janus-faced, however. While under physiological conditions iron-mediated reactive oxygen species generation boosts normal calcium-dependent signaling pathways, excessive iron levels promote oxidative stress leading to the upsurge of unrestrained calcium signals that damage mitochondrial function, among other downstream targets. Similarly, increases in mitochondrial calcium to non-physiological levels result in mitochondrial dysfunction and a predicted loss of iron homeostasis. Hence, if uncontrolled, the iron/calcium self-feeding cycle becomes deleterious to neuronal function, leading eventually to neuronal death. Here, we review the multiple cell-damaging responses generated by the unregulated iron/calcium self-feeding cycle, such as excitotoxicity, free radical-mediated lipid peroxidation, and the oxidative modification of crucial components of iron and calcium homeostasis/signaling: the iron transporter DMT1, plasma membrane, and intracellular calcium channels and pumps. We discuss also how iron-induced dysregulation of mitochondrial calcium contributes to the generation of neurodegenerative conditions, including Alzheimer’s disease (AD) and Parkinson’s disease (PD).
Collapse
Affiliation(s)
- Marco Tulio Núñez
- Iron and Neuroregeneration Laboratory, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Calcium Signaling Laboratory, Biomedical Research Institute, CEMC, Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
28
|
Cholangiocyte death in ductopenic cholestatic cholangiopathies: Mechanistic basis and emerging therapeutic strategies. Life Sci 2019; 218:324-339. [DOI: 10.1016/j.lfs.2018.12.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 12/26/2018] [Indexed: 02/07/2023]
|
29
|
Jung ME, Mallet RT. Intermittent hypoxia training: Powerful, non-invasive cerebroprotection against ethanol withdrawal excitotoxicity. Respir Physiol Neurobiol 2018; 256:67-78. [PMID: 28811138 PMCID: PMC5825251 DOI: 10.1016/j.resp.2017.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 07/24/2017] [Accepted: 08/08/2017] [Indexed: 12/12/2022]
Abstract
Ethanol intoxication and withdrawal exact a devastating toll on the central nervous system. Abrupt ethanol withdrawal provokes massive release of the excitatory neurotransmitter glutamate, which over-activates its postsynaptic receptors, causing intense Ca2+ loading, p38 mitogen activated protein kinase activation and oxidative stress, culminating in ATP depletion, mitochondrial injury, amyloid β deposition and neuronal death. Collectively, these mechanisms produce neurocognitive and sensorimotor dysfunction that discourages continued abstinence. Although the brain is heavily dependent on blood-borne O2 to sustain its aerobic ATP production, brief, cyclic episodes of moderate hypoxia and reoxygenation, when judiciously applied over the course of days or weeks, evoke adaptations that protect the brain from ethanol withdrawal-induced glutamate excitotoxicity, mitochondrial damage, oxidative stress and amyloid β accumulation. This review summarizes evidence from ongoing preclinical research that demonstrates intermittent hypoxia training to be a potentially powerful yet non-invasive intervention capable of affording robust, sustained neuroprotection during ethanol withdrawal.
Collapse
Affiliation(s)
- Marianna E Jung
- Center for Neuroscience Discovery, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107-2699, USA.
| | - Robert T Mallet
- Institute for Cardiovascular and Metabolic Diseases, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107-2699, USA.
| |
Collapse
|
30
|
Matei N, Camara R, Zhang JH. Emerging mechanisms and novel applications of hydrogen gas therapy. Med Gas Res 2018; 8:98-102. [PMID: 30319764 PMCID: PMC6178641 DOI: 10.4103/2045-9912.239959] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
Clinical and pre-clinical studies have reported a broad range of applications for hydrogen gas therapy. Classically, conventional antioxidant therapy is limited because it neutralizes both the detrimental and protective effects of reactive oxygen species. As a weak reducing agent, hydrogen gas avoids this paradox by reacting with strong oxidants while leaving other beneficial oxidants reactive. This review gathers a promising list of hydrogen gas applications that merit further mechanistic investigation and additional therapeutic trials. Reports support the ability of hydrogen gas to downregulate the expression of pro-inflammatory cytokines and pro-apoptotic factors. Mechanistically, hydrogen gas has been shown to downregulate miR-9 and miR-21, while upregulating miR-199 to reduce inflammatory injury. In angiogenic pathways, hydrogen's inhibition of cyclic guanosine monophosphate-degrading phosphodiesterase led to higher levels of cyclic guanosine monophosphate, activation of protein kinase, and angiogenesis; next, as hydrogen gas increased the levels of intracellular calcium, stimulated vascular endothelial growth factor increased nitric oxide production. In conjunction, hydrogen gas opened adenosine triphosphate-sensitive potassium channel channels, which activate downstream mitogen-activated protein kinase pathways. Growing molecular mechanisms have discovered a plethora of downstream targets for hydrogen gas therapy that include autophagy (via the adenosine 5’-monophosphate-activated protein kinase/mammalian target of rapamycin pathway), histone modification, mitochondrial unfolded protein response, acute oxidative stress after exercise, and oxidative stress secondary to aging. In conclusion, evolving research has discovered novel molecular connections that will continue to widen applications for hydrogen therapy.
Collapse
Affiliation(s)
- Nathanael Matei
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Richard Camara
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA.,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
31
|
Yu L, Scherlag BS, Dormer K, Rutel I, Huang B, Zhou X, Kuriakose AE, Nguyen KK, Po S. Targeted Ganglionated Plexi Denervation Using Magnetic Nanoparticles Carrying Calcium Chloride Payload. JACC Clin Electrophysiol 2018; 4:1347-1358. [PMID: 30336881 DOI: 10.1016/j.jacep.2018.06.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 11/28/2022]
Abstract
OBJECTIVES This study sought to develop a novel targeted delivery therapy to ablate the major atrial ganglionated plexi (GP) using magnetic nanoparticles carrying a CaCl2 payload. BACKGROUND Prior studies indicated the role of hyperactivity of the cardiac autonomic nervous system in the genesis of atrial fibrillation. METHODS Twenty-eight male mongrel dogs underwent a bilateral thoracotomy. CaCl2-encapsulated magnetic nanoparticles (Ca-MNP) included magnetite in a sphere of biocompatible, biodegradable poly(lactic-co-glycolic acid). A custom external electromagnet focusing the magnetic field gradient (2,600 G) on the epicardial surface of the targeted GP was used to pull Ca-MNP into and release CaCl2 within the GP. The ventricular rate slowing response to high frequency stimulation (20 Hz, 0.1 ms) of the GP was used to assess the GP function. RESULTS The minimal effective concentration of CaCl2 to inhibit the GP function was 0.5 mmol/l. Three weeks after CaCl2 (0.5 mmol/l, n = 18 GP) or saline (n = 18 GP) microinjection into GP, the increased GP function, neural activity, and atrial fibrillation inducibility, as well as shortened effective refractory period in response to 6 h of rapid atrial pacing (1,200 beats/min) were suppressed by CaCl2 microinjection. After intracoronary infusion of Ca-MNP, the external electromagnet pulled Ca-MNP to the targeted GP and suppressed the GP function (n = 6 GP) within 15 min. CONCLUSIONS Ca-MNP can be magnetically targeted to suppress GP function by calcium-mediated neurotoxicity. This novel approach may be used to treat arrhythmias related to hyperactivity of the cardiac autonomic nervous system, such as early stage of atrial fibrillation, with minimal myocardial injury.
Collapse
Affiliation(s)
- Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Benjamin S Scherlag
- Section of Cardiovascular Diseases and Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kenneth Dormer
- Integrative Physiology and Pharmacology Department, College of Osteopathic Medicine, Liberty University, Lynchburg, Virginia
| | - Isaac Rutel
- Department of Radiological Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Bing Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Aneetta E Kuriakose
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas
| | - Kytai K Nguyen
- Department of Bioengineering, the University of Texas at Arlington, Arlington, Texas
| | - Sunny Po
- Section of Cardiovascular Diseases and Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| |
Collapse
|
32
|
Bossio C, Abdel Aziz I, Tullii G, Zucchetti E, Debellis D, Zangoli M, Di Maria F, Lanzani G, Antognazza MR. Photocatalytic Activity of Polymer Nanoparticles Modulates Intracellular Calcium Dynamics and Reactive Oxygen Species in HEK-293 Cells. Front Bioeng Biotechnol 2018; 6:114. [PMID: 30211158 PMCID: PMC6119808 DOI: 10.3389/fbioe.2018.00114] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/20/2018] [Indexed: 11/13/2022] Open
Abstract
Optical modulation of living cells activity by light-absorbing exogenous materials is gaining increasing interest, due to the possibility both to achieve high spatial and temporal resolution with a minimally invasive and reversible technique and to avoid the need of viral transfection with light-sensitive proteins. In this context, conjugated polymers represent ideal candidates for photo-transduction, due to their excellent optoelectronic and biocompatibility properties. In this work, we demonstrate that organic polymer nanoparticles, based on poly(3-hexylthiophene) conjugated polymer, establish a functional interaction with an in vitro cell model (Human Embryonic Kidney cells, HEK-293). They display photocatalytic activity in aqueous environment and, once internalized within the cell cytosol, efficiently generate reactive oxygen species (ROS) upon visible light excitation, without affecting cell viability. Interestingly, light-activated ROS generation deterministically triggers modulation of intracellular calcium ion flux, successfully controlled at the single cell level. In perspective, the capability of polymer NPs to produce ROS and to modulate Ca2+ dynamics by illumination on-demand, at non-toxic levels, may open the path to the study of biological processes with a gene-less approach and unprecedented spatio-temporal resolution, as well as to the development of new biotechnology tools for cell optical modulation.
Collapse
Affiliation(s)
- Caterina Bossio
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
| | - Ilaria Abdel Aziz
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
- Dipartimento di Fisica, Politecnico di Milano, Milan, Italy
| | - Gabriele Tullii
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
- Dipartimento di Fisica, Politecnico di Milano, Milan, Italy
| | - Elena Zucchetti
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
- Dipartimento di Fisica, Politecnico di Milano, Milan, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, Genova, Italy
| | - Mattia Zangoli
- Institute for Organic Synthesis and Photoreactivity, CNR-ISOF, Bologna, Italy
| | - Francesca Di Maria
- Institute for Organic Synthesis and Photoreactivity, CNR-ISOF, Bologna, Italy
| | - Guglielmo Lanzani
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
- Dipartimento di Fisica, Politecnico di Milano, Milan, Italy
| | - Maria Rosa Antognazza
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
| |
Collapse
|
33
|
Kim H, Thompson J, Ji G, Ganapathy V, Neugebauer V. Monomethyl fumarate inhibits pain behaviors and amygdala activity in a rat arthritis model. Pain 2018; 158:2376-2385. [PMID: 28832396 DOI: 10.1097/j.pain.0000000000001042] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Neuroplasticity in the amygdala, a brain center for emotions, leads to increased neuronal activity and output that can generate emotional-affective behaviors and modulate nocifensive responses. Mechanisms of increased activity in the amygdala output region (central nucleus, CeA) include increased reactive oxygen species, and so we explored beneficial effects of monomethyl fumarate (MMF), which can have neuroprotective effects through the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) antioxidant response pathway. Systemic (intraperitoneal) MMF dose-dependently inhibited vocalizations and mechanosensitivity (hindlimb withdrawal reflexes) of rats in an arthritis pain model (kaolin-carrageenan-induced monoarthritis in the knee). Stereotaxic administration of MMF into the CeA by microdialysis also inhibited vocalizations but had a limited effect on mechanosensitivity, suggesting a differential contribution to emotional-affective vs sensory pain aspects. Extracellular single-unit recordings of CeA neurons in anesthetized rats showed that stereotaxic administration of MMF into the CeA by microdialysis inhibited background activity and responses of CeA neurons to knee joint stimulation in the arthritis pain model. Monomethyl fumarate had no effect on behaviors and neuronal activity under normal conditions. The results suggest that MMF can inhibit emotional-affective responses in an arthritis pain model through an action that involves the amygdala (CeA).
Collapse
Affiliation(s)
- Hyunyoung Kim
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center (TTUHSC), School of Medicine, Lubbock, TX, USA
| | - Jeremy Thompson
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center (TTUHSC), School of Medicine, Lubbock, TX, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center (TTUHSC), School of Medicine, Lubbock, TX, USA
| | - Vadivel Ganapathy
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center (TTUHSC), Lubbock, TX, USA.,Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center (TTUHSC), Lubbock, TX, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center (TTUHSC), School of Medicine, Lubbock, TX, USA.,Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center (TTUHSC), Lubbock, TX, USA
| |
Collapse
|
34
|
Kimura S, Iwasaki T, Oe K, Shimizu K, Suemori T, Kanazawa T, Shioji N, Kuroe Y, Matsuoka Y, Morimatsu H. High Ionized Calcium Concentration Is Associated With Prolonged Length of Stay in the Intensive Care Unit for Postoperative Pediatric Cardiac Patients. J Cardiothorac Vasc Anesth 2018; 32:1667-1675. [DOI: 10.1053/j.jvca.2017.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Indexed: 11/11/2022]
|
35
|
Anderson CJ, Kahl A, Qian L, Stepanova A, Starkov A, Manfredi G, Iadecola C, Zhou P. Prohibitin is a positive modulator of mitochondrial function in PC12 cells under oxidative stress. J Neurochem 2018; 146:235-250. [PMID: 29808474 PMCID: PMC6105506 DOI: 10.1111/jnc.14472] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/10/2018] [Accepted: 05/23/2018] [Indexed: 12/15/2022]
Abstract
Prohibitin (PHB) is a ubiquitously expressed and evolutionarily conserved mitochondrial protein with multiple functions. We have recently shown that PHB up-regulation offers robust protection against neuronal injury in models of cerebral ischemia in vitro and in vivo, but the mechanism by which PHB affords neuroprotection remains to be elucidated. Here, we manipulated PHB expression in PC12 neural cells to investigate its impact on mitochondrial function and the mechanisms whereby it protects cells exposed to oxidative stress. PHB over-expression promoted cell survival, whereas PHB down-regulation diminished cell viability. Functionally, manipulation of PHB levels did not affect basal mitochondrial respiration, but it increased spare respiratory capacity. Moreover, PHB over-expression preserved mitochondrial respiratory function of cells exposed to oxidative stress. Preserved respiratory capacity in differentiated PHB over-expressing cells exposed to oxidative stress was associated with an elongated mitochondrial morphology, whereas PHB down-regulation enhanced fragmentation. Mitochondrial complex I oxidative degradation was attenuated by PHB over-expression and increased in PHB knockdown cells. Changes in complex I degradation were associated with alterations of respiratory chain supercomplexes. Furthermore, we showed that PHB directly interacts with cardiolipin and that down-regulation of PHB results in loss of cardiolipin in mitochondria, which may contribute to destabilizing respiratory chain supercomplexes. Taken together, these data demonstrate that PHB modulates mitochondrial integrity and bioenergetics under oxidative stress, and suggest that the protective effect of PHB is mediated by stabilization of the mitochondrial respiratory machinery and its functional capacity, by the regulation of cardiolipin content. Open Data: Materials are available on https://cos.io/our-services/open-science-badges/ https://osf.io/93n6m/.
Collapse
Affiliation(s)
| | | | - Liping Qian
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61 Street, New York, NY 10065
| | - Anna Stepanova
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61 Street, New York, NY 10065
| | - Anatoly Starkov
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61 Street, New York, NY 10065
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61 Street, New York, NY 10065
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61 Street, New York, NY 10065
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61 Street, New York, NY 10065
| |
Collapse
|
36
|
Oxidative stress, caspase-3 activation and cleavage of ROCK-1 play an essential role in MeHg-induced cell death in primary astroglial cells. Food Chem Toxicol 2018; 113:328-336. [PMID: 29428217 DOI: 10.1016/j.fct.2018.01.057] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 12/13/2022]
Abstract
Methylmercury is a toxic environmental contaminant that elicits significant toxicity in humans. The central nervous system is the primary target of toxicity, and is particularly vulnerable during development. Rho-associated protein kinase 1 (ROCK-1) is a major downstream effector of the small GTPase RhoA and a direct substrate of caspase-3. The activation of ROCK-1 is necessary for membrane blebbing during apoptosis. In this work, we examined whether MeHg could affect the RhoA/ROCK-1 signaling pathway in primary cultures of mouse astrocytes. Exposure of cells with 10 μM MeHg decreased cellular viability after 24 h of incubation. This reduction in viability was preceded by a significant increase in intracellular and mitochondrial reactive oxygen species levels, as well as a reduced NAD+/NADH ratio. MeHg also induced an increase in mitochondrial-dependent caspase-9 and caspase-3, while the levels of RhoA protein expression were reduced or unchanged. We further found that MeHg induced ROCK-1 cleavage/activation and promoted LIMK1 and MYPT1 phosphorylation, both of which are the best characterized ROCK-1 downstream targets. Inhibiting ROCK-1 and caspases activation attenuated the MeHg-induced cell death. Collectively, these findings are the first to show that astrocytes exposed to MeHg showed increased cleavage/activation of ROCK-1, which was independent of the small GTPase RhoA.
Collapse
|
37
|
Xie Y, Wanggou S, Liu Q, Li X, Liu J, Wu M. A brain-specific isoform of apoptosis-inducing factor 2 attenuates ischemia-induced oxidative stress in HT22 cells. Neurochem Int 2018; 112:179-186. [DOI: 10.1016/j.neuint.2017.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/25/2017] [Accepted: 07/16/2017] [Indexed: 11/30/2022]
|
38
|
Chronic Administration of S-Allylcysteine Activates Nrf2 Factor and Enhances the Activity of Antioxidant Enzymes in the Striatum, Frontal Cortex and Hippocampus. Neurochem Res 2017. [PMID: 28646259 DOI: 10.1007/s11064-017-2337-2] [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: 10/19/2022]
Abstract
Oxidative stress plays an important role in neurodegenerative diseases and aging. The cellular defense mechanisms to deal with oxidative damage involve the activation of transcription factor related to NF-E2 (Nrf2), which enhances the transcription of antioxidant and phase II enzyme genes. S-allylcysteine (SAC) is an antioxidant with neuroprotective properties, and the main organosulfur compound in aged garlic extract. The ability of SAC to activate the Nrf2 factor has been previously reported in hepatic cells; however this effect has not been studied in normal brain. In order to determine if the chronic administration of SAC is able to activate Nrf2 factor and enhance antioxidant defense in the brain, male Wistar rats were administered with SAC (25, 50, 100 and 200 mg/kg-body weight each 24 h, i.g.) for 90 days. The activation of Nrf2, the levels of p65 and 8-hydroxy-2-deoxyguanosine (8-OHdG) as well as the activities of the enzymes glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT), superoxide dismutase (SOD), and glutathione S-transferase (GST) were evaluated in the hippocampus, striatum and frontal cortex. Results showed that SAC activated Nrf2 factor in the hippocampus (25-200 mg/kg) and striatum (100 mg/kg) and significantly decreased p65 levels in the frontal cortex (25-200 mg/kg). On the other hand, SAC increased GPx, GR, CAT and SOD activities mainly in the hippocampus and striatum, but it did not change GST activity. Finally, no changes were observed in 8-OHdG levels mediated by SAC in any brain region, but the hippocampus showed a major level of 8-OHdG compared with the striatum and frontal cortex. All these results suggest that in the hippocampus, the observed increase in the activity of antioxidant enzymes could be associated with the ability of SAC to activate Nrf2 factor; however, a different mechanism could be involved in the striatum and frontal cortex, since no changes were found in Nrf2 activation and p65 levels.
Collapse
|
39
|
Lee JC, Kim YH, Lee TK, Kim IH, Cho JH, Cho GS, Shin BN, Park JH, Ahn JH, Shin MC, Cho JH, Kang IJ, Won MH, Seo JY. Effects of ischemic preconditioning on PDGF-BB expression in the gerbil hippocampal CA1 region following transient cerebral ischemia. Mol Med Rep 2017. [PMID: 28627606 PMCID: PMC5562056 DOI: 10.3892/mmr.2017.6799] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Ischemic preconditioning (IPC) is induced by exposure to brief durations of transient ischemia, which results in ischemic tolerance to a subsequent longer or lethal period of ischemia. In the present study, the effects of IPC (2 min of transient cerebral ischemia) were examined on immunoreactivity of platelet‑derived growth factor (PDGF)‑BB and on neuroprotection in the gerbil hippocampal CA1 region following lethal transient cerebral ischemia (LTCI; 5 min of transient cerebral ischemia). IPC was subjected to a 2‑min sublethal ischemia and a LTCI was given 5‑min transient ischemia. The animals in all of the groups were given recovery times of 1, 2 and 5 days and change in PDGF‑BB immunoreactivity was examined as was the neuronal damage/death in the hippocampus induced by LTCI. LTCI induced a significant loss of pyramidal neurons in the hippocampal CA1 region 5 days after LTCI, and significantly decreased PDGF‑BB immunoreactivity in the CA1 pyramidal neurons from day 1 after LTCI. Conversely, IPC effectively protected the CA1 pyramidal neurons from LTCI and increased PDGF‑BB immunoreactivity in the CA1 pyramidal neurons post‑LTCI. In conclusion, the results demonstrated that LTCI significantly altered PDGF‑BB immunoreactivity in pyramidal neurons in the hippocampal CA1 region, whereas IPC increased the immunoreactivity. These findings indicated that PDGF‑BB may be associated with IPC‑mediated neuroprotection.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yang Hee Kim
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Geum-Sil Cho
- Pharmacology and Toxicology Department, Shinpoong Pharmaceutical Co., Ltd., Ansan, Gyeonggi 15610, Republic of Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jeong Yeol Seo
- Department of Emergency Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| |
Collapse
|
40
|
Sasaki T, Awaji T, Shimada K, Sasaki H. Increase of reactive oxygen species generation in cerebral cortex slices after the transiently enhanced metabolic activity. Neurosci Res 2017; 123:55-64. [PMID: 28499835 DOI: 10.1016/j.neures.2017.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/31/2017] [Accepted: 04/28/2017] [Indexed: 10/19/2022]
Abstract
Under certain conditions such as hypoxia-reoxygenation, the generation of reactive oxygen species (ROS) increases following hypoxia caused by a decreased oxygen supply. As another hypoxic condition, an excess neural activity status including epileptic seizure induces a decrease in tissue oxygen partial pressure (pO2) caused by enhanced oxygen utilization; however, whether ROS generation increases following the hypoxic status induced by transiently enhanced energy metabolism in brain tissue currently remains unknown. We herein investigated ROS-dependent chemiluminescence in cerebral cortex slices during the restoration of transiently enhanced energy metabolism induced by a high-potassium treatment with tissue pO2 changes and redox balance. ROS generation in the tissue was enhanced after high-potassium-induced hypoxia, but not by the reversed order of the treatment: control-potassium then high-potassium treatment, high-potassium treatment alone, and control-potassium treatment alone. The high-potassium treatment induced a transient decrease in tissue pO2 and a shift in the tissue redox balance towards reduction. The transient shift in the tissue redox balance towards reduction with enhanced metabolic activity and its recovery may correlate with ROS generation. This phenomenon may mimic ROS generation following the hypoxic status induced by transiently enhanced energy metabolism.
Collapse
Affiliation(s)
- Toru Sasaki
- Department of Medical Engineering and Technology, Kitasato University School of Allied of Health Sciences, 1-15-1 Kitasato, Sagamihara, Kanagawa 252-0373, Japan; Research Team for Mechanism of Aging, Redox Research, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi, Tokyo 173-0015, Japan.
| | - Takuji Awaji
- Department of Medical Engineering and Technology, Kitasato University School of Allied of Health Sciences, 1-15-1 Kitasato, Sagamihara, Kanagawa 252-0373, Japan
| | - Kazuyoshi Shimada
- Department of Medical Engineering and Technology, Kitasato University School of Allied of Health Sciences, 1-15-1 Kitasato, Sagamihara, Kanagawa 252-0373, Japan
| | - Haruyo Sasaki
- Department of Medical Engineering and Technology, Kitasato University School of Allied of Health Sciences, 1-15-1 Kitasato, Sagamihara, Kanagawa 252-0373, Japan
| |
Collapse
|
41
|
Spät A, Szanda G. The Role of Mitochondria in the Activation/Maintenance of SOCE: Store-Operated Ca 2+ Entry and Mitochondria. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:257-275. [PMID: 28900919 DOI: 10.1007/978-3-319-57732-6_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mitochondria extensively modify virtually all cellular Ca2+ transport processes, and store-operated Ca2+ entry (SOCE) is no exception to this rule. The interaction between SOCE and mitochondria is complex and reciprocal, substantially altering and, ultimately, fine-tuning both capacitative Ca2+ influx and mitochondrial function. Mitochondria, owing to their considerable Ca2+ accumulation ability, extensively buffer the cytosolic Ca2+ in their vicinity. In turn, the accumulated ion is released back into the neighboring cytosol during net Ca2+ efflux. Since store depletion itself and the successive SOCE are both Ca2+-regulated phenomena, mitochondrial Ca2+ handling may have wide-ranging effects on capacitative Ca2+ influx at any given time. In addition, mitochondria may also produce or consume soluble factors known to affect store-operated channels. On the other hand, Ca2+ entering the cell during SOCE is sensed by mitochondria, and the ensuing mitochondrial Ca2+ uptake boosts mitochondrial energy metabolism and, if Ca2+ overload occurs, may even lead to apoptosis or cell death. In several cell types, mitochondria seem to be sterically excluded from the confined space that forms between the plasma membrane (PM) and endoplasmic reticulum (ER) during SOCE. This implies that high-Ca2+ microdomains comparable to those observed between the ER and mitochondria do not form here. In the following chapter, the above aspects of the many-sided SOCE-mitochondrion interplay will be discussed in greater detail.
Collapse
Affiliation(s)
- András Spät
- Department of Physiology, Semmelweis University Medical School, POB 2, 1428, Budapest, Hungary.
- Laboratory of Molecular Physiology, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Gergö Szanda
- Department of Physiology, Semmelweis University Medical School, POB 2, 1428, Budapest, Hungary
| |
Collapse
|
42
|
Kučera O, Endlicher R, Rychtrmoc D, Lotková H, Sobotka O, Červinková Z. Acetaminophen toxicity in rat and mouse hepatocytes in vitro. Drug Chem Toxicol 2016; 40:448-456. [PMID: 27960556 DOI: 10.1080/01480545.2016.1255953] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CONTEXT Acetaminophen (APAP) hepatotoxicity is often studied in primary cultures of hepatocytes of various species, but there are only few works comparing interspecies differences in susceptibility of hepatocytes to APAP in vitro. OBJECTIVES The aim of our work was to compare hepatotoxicity of APAP in rat and mouse hepatocytes in primary cultures. MATERIALS AND METHODS Hepatocytes isolated from male Wistar rats and C57Bl/6J mice were exposed to APAP for up to 24 h. We determined lactate dehydrogenase (LDH) activity in culture medium, activity of cellular dehydrogenases (WST-1) and activity of caspases 3 in cell lysate as markers of cell damage/death. We assessed content of intracellular reduced glutathione, production of reactive oxygen species (ROS) and malondialdehyde (MDA). Respiration of digitonin-permeabilized hepatocytes was measured by high resolution respirometry and mitochondrial membrane potential (MMP) was visualized (JC-1). RESULTS APAP from concentrations of 2.5 and 0.75 mmol/L induced a decrease in viability of rat (p < 0.001) and mouse (p < 0.001) hepatocytes (WST-1), respectively. In contrast to rat hepatocytes, there was no activation of caspase-3 in mouse hepatocytes after APAP treatment. Earlier damage to plasma membrane and faster depletion of reduced glutathione were detected in mouse hepatocytes. Mouse hepatocytes showed increased glutamate + malate-driven respiration in state 4 and higher susceptibility of the outer mitochondrial membrane (OMM) to APAP-induced injury. CONCLUSION APAP displayed dose-dependent toxicity in hepatocytes of both species. Mouse hepatocytes in primary culture however had approximately three-fold higher susceptibility to the toxic effect of APAP when compared to rat hepatocytes.
Collapse
Affiliation(s)
| | - René Endlicher
- b Department of Anatomy , Charles University in Prague, Faculty of Medicine in Hradec Králové , Hradec Králové , Czech Republic
| | | | | | | | | |
Collapse
|
43
|
Rhoney DH, Parker D. Considerations in Fluids and Electrolytes After Traumatic Brain Injury. Nutr Clin Pract 2016; 21:462-78. [PMID: 16998145 DOI: 10.1177/0115426506021005462] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Appropriate fluid management of patients with traumatic brain injury (TBI) presents a challenge for many clinicians. Many of these patients may receive osmotic diuretics for the treatment of increased intracranial pressure or develop sodium disturbances, which act to alter fluid balance. However, establishment of fluid balance is extremely important for improving patient outcomes after neurologic injury. The use of hyperosmolar fluids, such as hypertonic saline, has gained significant interest because they are devoid of dehydrating properties and may have other beneficial properties for patients with TBI. Electrolyte derangements are also common after neurologic injury, with many having neurologic manifestations. In addition, the role of electrolyte abnormalities in the secondary neurologic injury cascade is being delineated and may offer a potential future therapeutic intervention.
Collapse
Affiliation(s)
- Denise H Rhoney
- Department of Pharmacy Practice, Wayne State University, Eugene Applebaum College of Pharmacy & Health Sciences, 259 Mack Avenue, Detroit, MI 48201, USA.
| | | |
Collapse
|
44
|
Hong Z, Tian Y, Yuan Y, Qi M, Li Y, Du Y, Chen L, Chen L. Enhanced Oxidative Stress Is Responsible for TRPV4-Induced Neurotoxicity. Front Cell Neurosci 2016; 10:232. [PMID: 27799895 PMCID: PMC5065954 DOI: 10.3389/fncel.2016.00232] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/26/2016] [Indexed: 11/20/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) has been reported to be responsible for neuronal injury in pathological conditions. Excessive oxidative stress can lead to neuronal damage, and activation of TRPV4 increases the production of reactive oxygen species (ROS) and nitric oxide (NO) in many types of cells. The present study explored whether TRPV4-induced neuronal injury is mediated through enhancing oxidative stress. We found that intracerebroventricular injection of the TRPV4 agonist GSK1016790A increased the content of methane dicarboxylic aldehyde (MDA) and NO in the hippocampus, which was blocked by administration of the TRPV4 specific antagonist HC-067047. The activities of catalase (CAT) and glutathione peroxidase (GSH-Px) were decreased by GSK1016790A, whereas the activity of superoxide dismutase (SOD) remained unchanged. Moreover, the protein level and activity of neuronal nitric oxide synthase (nNOS) were increased by GSK1016790A, and the GSK1016790A-induced increase in NO content was blocked by an nNOS specific antagonist ARL-17477. The GSK1016790A-induced modulations of CAT, GSH-Px and nNOS activities and the protein level of nNOS were significantly inhibited by HC-067047. Finally, GSK1016790A-induced neuronal death and apoptosis in the hippocampal CA1 area were markedly attenuated by administration of a ROS scavenger Trolox or ARL-17477. We conclude that activation of TRPV4 enhances oxidative stress by inhibiting CAT and GSH-Px and increasing nNOS, which is responsible, at least in part, for TRPV4-induced neurotoxicity.
Collapse
Affiliation(s)
- Zhiwen Hong
- Department of Physiology, Nanjing Medical University Nanjing, China
| | - Yujing Tian
- Department of Physiology, Nanjing Medical University Nanjing, China
| | - Yibiao Yuan
- The Laboratory Center for Basic Medical Sciences, Nanjing Medical University Nanjing, China
| | - Mengwen Qi
- Department of Physiology, Nanjing Medical University Nanjing, China
| | - Yingchun Li
- Department of Physiology, Nanjing Medical University Nanjing, China
| | - Yimei Du
- Research Center of Ion Channelopathy, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Lei Chen
- Department of Physiology, Nanjing Medical University Nanjing, China
| | - Ling Chen
- Department of Physiology, Nanjing Medical University Nanjing, China
| |
Collapse
|
45
|
Radak Z, Suzuki K, Higuchi M, Balogh L, Boldogh I, Koltai E. Physical exercise, reactive oxygen species and neuroprotection. Free Radic Biol Med 2016; 98:187-196. [PMID: 26828019 DOI: 10.1016/j.freeradbiomed.2016.01.024] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/13/2016] [Accepted: 01/28/2016] [Indexed: 12/17/2022]
Abstract
Regular exercise has systemic beneficial effects, including the promotion of brain function. The adaptive response to regular exercise involves the up-regulation of the enzymatic antioxidant system and modulation of oxidative damage. Reactive oxygen species (ROS) are important regulators of cell signaling. Exercise, via intensity-dependent modulation of metabolism and/or directly activated ROS generating enzymes, regulates the cellular redox state of the brain. ROS are also involved in the self-renewal and differentiation of neuronal stem cells and the exercise-mediated neurogenesis could be partly associated with ROS production. Exercise has strong effects on the immune system and readily alters the production of cytokines. Certain cytokines, especially IL-6, IL-1, TNF-α, IL-18 and IFN gamma, are actively involved in the modulation of synaptic plasticity and neurogenesis. Cytokines can also contribute to ROS production. ROS-mediated alteration of lipids, protein, and DNA could directly affect brain function, while exercise modulates the accumulation of oxidative damage. Oxidative alteration of macromolecules can activate signaling processes, membrane remodeling, and gene transcription. The well known neuroprotective effects of exercise are partly due to redox-associated adaptation.
Collapse
Affiliation(s)
- Zsolt Radak
- Institute of Sport Science, University of Physical Education, Alkotas u. 44, TF, Budapest, Hungary; Graduate School of Sport Sciences, Waseda University, Saitama, Japan.
| | - Katsuhiko Suzuki
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Mitsuru Higuchi
- Graduate School of Sport Sciences, Waseda University, Saitama, Japan
| | - Laszlo Balogh
- Institute of Physical Education and Sport Science, University of Szeged, Hungary
| | - Istvan Boldogh
- Department of Microbiology and Immunology, Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Erika Koltai
- Institute of Sport Science, University of Physical Education, Alkotas u. 44, TF, Budapest, Hungary
| |
Collapse
|
46
|
Chang JC, Wu SL, Liu KH, Chen YH, Chuang CS, Cheng FC, Su HL, Wei YH, Kuo SJ, Liu CS. Allogeneic/xenogeneic transplantation of peptide-labeled mitochondria in Parkinson's disease: restoration of mitochondria functions and attenuation of 6-hydroxydopamine-induced neurotoxicity. Transl Res 2016; 170:40-56.e3. [PMID: 26730494 DOI: 10.1016/j.trsl.2015.12.003] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/03/2015] [Accepted: 12/03/2015] [Indexed: 01/31/2023]
Abstract
Although restoration of mitochondrial function in mitochondrial diseases through peptide-mediated allogeneic mitochondrial delivery (PMD) has been demonstrated in vitro, the in vivo therapeutic efficacy of PMD in Parkinson's disease (PD) has yet to be determined. In this study, we compared the functionality of mitochondrial transfer with or without Pep-1 conjugation in neurotoxin (6-hydroxydopamine, 6-OHDA)-induced PC12 cells and PD rat models. We injected mitochondria into the medial forebrain bundle (MFB) of the PD rats after subjecting the nigrostriatal pathway to a unilateral 6-OHDA lesion for 21 days, and we verified the effectiveness of the mitochondrial graft in enhancing mitochondrial function in the soma of the substantia nigra (SN) neuron through mitochondrial transport dynamics in the nigrostriatal circuit. The result demonstrated that only PMD with allogeneic and xenogeneic sources significantly sustained mitochondrial function to resist the neurotoxin-induced oxidative stress and apoptotic death in the rat PC12 cells. The remaining cells exhibited a greater capability of neurite outgrowth. Furthermore, allogeneic and xenogeneic transplantation of peptide-labeled mitochondria after 3 months improved the locomotive activity in the PD rats. This increase was accompanied by a marked decrease in dopaminergic neuron loss in the substantia nigra pars compacta (SNc) and consistent enhancement of tyrosine hydroxylase-positive immunoreaction of dopaminergic neurons in the SNc and striatum. We also observed that in the SN dopaminergic neuron in the treated PD rats, mitochondrial complex I protein and mitochondrial dynamics were restored, thus ameliorating the oxidative DNA damage. Moreover, we determined signal translocation of graft allogeneic mitochondria from the MFB to the calbindin-positive SN neuron, which demonstrated the regulatory role of mitochondrial transport in alleviating 6-OHDA-induced degeneration of dopaminergic neurons.
Collapse
Affiliation(s)
- Jui-Chih Chang
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Shey-Lin Wu
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| | - Ko-Hung Liu
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ya-Hui Chen
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Chieh-Sen Chuang
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| | - Fu-Chou Cheng
- Department of Medical Research, Stem Cell Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Yau-Huei Wei
- Department of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan; Department of Medicine, Mackay Medical College, Taipei, Taiwan
| | - Shou-Jen Kuo
- Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Chin-San Liu
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan; Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
47
|
Lu YF, Neugebauer V, Chen J, Li Z. Distinct contributions of reactive oxygen species in amygdala to bee venom-induced spontaneous pain-related behaviors. Neurosci Lett 2016; 619:68-72. [PMID: 26971700 DOI: 10.1016/j.neulet.2016.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/25/2016] [Accepted: 03/07/2016] [Indexed: 10/22/2022]
Abstract
Reactive oxygen species (ROS), such as superoxide and hydrogen peroxide, play essential roles in physiological plasticity and are also involved in the pathogenesis of persistent pain. Roles of peripheral and spinal ROS in pain have been well established, but much less is known about ROS in the amygdala, a brain region that plays an important role in pain modulation. The present study explored the contribution of ROS in the amygdala to bee venom (BV)-induced pain behaviors. Our data show that the amygdala is activated following subcutaneous BV injection into the left hindpaw, which is reflected in the increased number of c-Fos positive cells in the central and basolateral amygdala nuclei in the right hemisphere. Stereotaxic administration of a ROS scavenger (tempol, 10mM), NADPH oxidase inhibitor (baicalein, 5mM) or lipoxygenase inhibitor (apocynin, 10mM) into the right amygdala attenuated the BV-induced spontaneous licking and lifting behaviors, but had no effect on BV-induced paw flinch reflexes. Our study provides further evidence for the involvement of the amygdala in nociceptive processing and pain behaviors, and that ROS in amygdala may be a potential target for treatment strategies to inhibit pain.
Collapse
Affiliation(s)
- Yun-Fei Lu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China; Key Laboratory of Brain Stress and Behavior, PLA, Xi'an 710038, PR China
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center (TTUHSC), Lubbock, TX, USA.
| | - Jun Chen
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China; Key Laboratory of Brain Stress and Behavior, PLA, Xi'an 710038, PR China; Beijing Institute for Brain Disorders, Beijing 100069, PR China
| | - Zhen Li
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China; Key Laboratory of Brain Stress and Behavior, PLA, Xi'an 710038, PR China.
| |
Collapse
|
48
|
Abstract
Accumulating evidence suggests an important contribution of reactive oxygen species (ROS) to pain and neuropsychiatric disorders, but their role in pain-related plasticity in the brain is largely unknown. Neuroplasticity in the central nucleus of the amygdala (CeA) correlates positively with pain behaviors in different models. Little is known, however, about mechanisms of visceral pain-related amygdala changes. The electrophysiological and behavioral studies reported here addressed the role of ROS in the CeA in a visceral pain model induced by intracolonic zymosan. Vocalizations to colorectal distension and anxiety-like behavior increased after intracolonic zymosan and were inhibited by intra-CeA application of a ROS scavenger (tempol, a superoxide dismutase mimetic). Tempol also induced a place preference in zymosan-treated rats but not in controls. Single-unit recordings of CeA neurons in anesthetized rats showed increases of background activity and responses to visceral stimuli after intracolonic zymosan. Intra-CeA application of tempol inhibited the increased activity but had no effect under normal conditions. Whole-cell patch-clamp recordings of CeA neurons in brain slices from zymosan-treated rats showed that tempol decreased neuronal excitability and excitatory synaptic transmission of presumed nociceptive inputs from the brainstem (parabrachial area) through a combination of presynaptic and postsynaptic actions. Tempol had no effect in brain slices from sham controls. The results suggest that ROS contribute to visceral pain-related hyperactivity of amygdala neurons and amygdala-dependent behaviors through a mechanism that involves increased excitatory transmission and excitability of CeA neurons.
Collapse
|
49
|
Evaluation of the effects of different supplementation on oxidative status in patients with rheumatoid arthritis. Clin Rheumatol 2016; 35:1909-1915. [DOI: 10.1007/s10067-016-3168-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 12/03/2015] [Accepted: 01/04/2016] [Indexed: 02/07/2023]
|
50
|
Ion Channels and Oxidative Stress as a Potential Link for the Diagnosis or Treatment of Liver Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3928714. [PMID: 26881024 PMCID: PMC4736365 DOI: 10.1155/2016/3928714] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/22/2015] [Accepted: 10/27/2015] [Indexed: 02/06/2023]
Abstract
Oxidative stress results from a disturbed balance between oxidation and antioxidant systems. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) may be either harmful or beneficial to the cells. Ion channels are transmembrane proteins that participate in a large variety of cellular functions and have been implicated in the development of a variety of diseases. A significant amount of the available drugs in the market targets ion channels. These proteins have sulfhydryl groups of cysteine and methionine residues in their structure that can be targeted by ROS and RNS altering channel function including gating and conducting properties, as well as the corresponding signaling pathways associated. The regulation of ion channels by ROS has been suggested to be associated with some pathological conditions including liver diseases. This review focuses on understanding the role and the potential association of ion channels and oxidative stress in liver diseases including fibrosis, alcoholic liver disease, and cancer. The potential association between ion channels and oxidative stress conditions could be used to develop new treatments for major liver diseases.
Collapse
|