1
|
Momin A, Perrotti S, Waldman SD. The role of mitochondrial reactive oxygen species in chondrocyte mechanotransduction. J Orthop Res 2024; 42:628-637. [PMID: 37804213 DOI: 10.1002/jor.25709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 10/09/2023]
Abstract
Chondrocytes are mechanosensitive cells able to sense and respond to external mechanical stimuli through the process of mechanotransduction. Previous studies have demonstrated that mechanical stimulation causes mitochondrial deformation leading to mitochondrial reactive oxygen species (ROS) release in a dose-dependent manner. For this reason, we focused on elucidating the role of mitochondrial ROS as anabolic signaling molecules in chondrocyte mechanotransduction. Chondrocyte-seeded agarose gels were subjected to mechanical stimuli and the effect on matrix synthesis, ROS production, and mitogen-activated protein kinases (MAPK) signaling was evaluated. Through the use of ROS-specific staining, superoxide anion was the primary ROS released in response to mechanical stimuli. The anabolic effect of mechanical stimulation was abolished in the presence of electron transport chain inhibitors (complexes I, III, and V) and superoxide anion scavengers. Subsequent studies were centered on the involvement of MAPK pathways (ERK1/2, p38, and JNK) in the mechanotransduction cascade. While disruption of the ERK1/2 pathway had no apparent effect, the anabolic effect of mechanical stimulation was abolished in the presence of p38 and JNK pathway inhibitors. This suggest the involvement of apoptosis stimulating kinase 1 (ASK1), an upstream redox-sensitive MAP3K shared by both the JNK and p38 pathways. Future experiments will focus on the involvement of the thioredoxin-ASK1 complex which disassociates in the presence of oxidative stress, allowing ASK1 to phosphorylate several MAP2Ks. Overall, these findings indicate superoxide anion as the primary ROS released in response to mechanical stimuli and that the resulting anabolic effect on chondrogenic matrix biosynthesis arises from the ROS-dependent activation of the p38 and JNK MAPKs.
Collapse
Affiliation(s)
- Aisha Momin
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Simona Perrotti
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Stephen D Waldman
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Farzipour S, Zefrei FJ, Bahadorikhalili S, Alvandi M, Salari A, Shaghaghi Z. Nanotechnology Utilizing Ferroptosis Inducers in Cancer Treatment. Anticancer Agents Med Chem 2024; 24:571-589. [PMID: 38275050 DOI: 10.2174/0118715206278427231215111526] [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: 10/06/2023] [Revised: 11/11/2023] [Accepted: 11/20/2023] [Indexed: 01/27/2024]
Abstract
Current cancer treatment options have presented numerous challenges in terms of reaching high efficacy. As a result, an immediate step must be taken to create novel therapies that can achieve more than satisfying outcomes in the fight against tumors. Ferroptosis, an emerging form of regulated cell death (RCD) that is reliant on iron and reactive oxygen species, has garnered significant attention in the field of cancer therapy. Ferroptosis has been reported to be induced by a variety of small molecule compounds known as ferroptosis inducers (FINs), as well as several licensed chemotherapy medicines. These compounds' low solubility, systemic toxicity, and limited capacity to target tumors are some of the significant limitations that have hindered their clinical effectiveness. A novel cancer therapy paradigm has been created by the hypothesis that ferroptosis induced by nanoparticles has superior preclinical properties to that induced by small drugs and can overcome apoptosis resistance. Knowing the different ideas behind the preparation of nanomaterials that target ferroptosis can be very helpful in generating new ideas. Simultaneously, more improvement in nanomaterial design is needed to make them appropriate for therapeutic treatment. This paper first discusses the fundamentals of nanomedicine-based ferroptosis to highlight the potential and characteristics of ferroptosis in the context of cancer treatment. The latest study on nanomedicine applications for ferroptosis-based anticancer therapy is then highlighted.
Collapse
Affiliation(s)
- Soghra Farzipour
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Jalali Zefrei
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeed Bahadorikhalili
- Department of Electronic Engineering, Universitat Rovira i Virgili, 43007, Tarragona, Spain
| | - Maryam Alvandi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Shaghaghi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
3
|
Pandey S, Mangmool S, Madreiter-Sokolowski CT, Wichaiyo S, Luangmonkong T, Parichatikanond W. Exendin-4 protects against high glucose-induced mitochondrial dysfunction and oxidative stress in SH-SY5Y neuroblastoma cells through GLP-1 receptor/Epac/Akt signaling. Eur J Pharmacol 2023:175896. [PMID: 37391007 DOI: 10.1016/j.ejphar.2023.175896] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/16/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023]
Abstract
Mitochondrial dysfunction under diabetic condition leads to the development and progression of neurodegenerative complications. Recently, the beneficial effects of glucagon-like peptide-1 (GLP-1) receptor agonists on diabetic neuropathies have been widely recognized. However, molecular mechanisms underlying the neuroprotective effects of GLP-1 receptor agonists against high glucose (HG)-induced neuronal damages is not completely elucidated. Here, we investigated the underlying mechanisms of GLP-1 receptor agonist treatment against oxidative stress, mitochondrial dysfunction, and neuronal damages under HG-conditions mimicking a diabetic hyperglycemic state in SH-SY5Y neuroblastoma cells. We revealed that treatment with exendin-4, a GLP-1 receptor agonist, not only increased the expression of survival markers, phospho-Akt/Akt and Bcl-2, but also decreased the expression of pro-apoptotic marker, Bax, and reduced the levels of reactive oxygen species (ROS) defense markers (catalase, SOD-2, and HO-1) under HG conditions. The expressions of mitochondrial function associated genes, MCU and UCP3, and mitochondrial fission genes, DRP1 and FIS1, were decreased by exendin-4 compared to non-treated levels, while the protein expression levels of mitochondrial homeostasis regulators, Parkin and PINK1, were enhanced. In addition, blockade of Epac and Akt activities was able to antagonize these neuroprotective effects of exendin-4. Collectively, we demonstrated that stimulation of GLP-1 receptor propagates a neuroprotective cascade against the oxidative stresses and mitochondrial dysfunctions as well as augments survival through the Epac/Akt-dependent pathway. Therefore, the revealed mechanisms underlying GLP-1 receptor pathway by preserving mitochondrial homeostasis would be a therapeutic candidate to alleviate neuronal dysfunctions and delay the progression of diabetic neuropathies.
Collapse
Affiliation(s)
- Sudhir Pandey
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Corina T Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, 8010, Austria
| | - Surasak Wichaiyo
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | - Theerut Luangmonkong
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, 10400, Thailand
| | | |
Collapse
|
4
|
Begum HM, Shen K. Intracellular and microenvironmental regulation of mitochondrial membrane potential in cancer cells. WIREs Mech Dis 2023; 15:e1595. [PMID: 36597256 PMCID: PMC10176868 DOI: 10.1002/wsbm.1595] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023]
Abstract
Cancer cells have an abnormally high mitochondrial membrane potential (ΔΨm ), which is associated with enhanced invasive properties in vitro and increased metastases in vivo. The mechanisms underlying the abnormal ΔΨm in cancer cells remain unclear. Research on different cell types has shown that ΔΨm is regulated by various intracellular mechanisms such as by mitochondrial inner and outer membrane ion transporters, cytoskeletal elements, and biochemical signaling pathways. On the other hand, the role of extrinsic, tumor microenvironment (TME) derived cues in regulating ΔΨm is not well defined. In this review, we first summarize the existing literature on intercellular mechanisms of ΔΨm regulation, with a focus on cancer cells. We then offer our perspective on the different ways through which the microenvironmental cues such as hypoxia and mechanical stresses may regulate cancer cell ΔΨm . This article is categorized under: Cancer > Environmental Factors Cancer > Biomedical Engineering Cancer > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Hydari Masuma Begum
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Keyue Shen
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
- USC Stem Cell, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
5
|
Sardoiwala MN, Mohanbhai SJ, Karmakar S, Choudhury SR. Hytrin loaded polydopamine-serotonin nanohybrid induces IDH2 mediated neuroprotective effect to alleviate Parkinson's disease. BIOMATERIALS ADVANCES 2022; 133:112602. [PMID: 35527145 DOI: 10.1016/j.msec.2021.112602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/12/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
Parkinson's disease (PD) is the second most neurodegenerative disease caused due to synucleinopathy leads to the death of dopaminergic and serotonergic neurons. The approach to reduce synucleinopathy paves the therapeutic way in PD management. Recent studies highlight anti-Parkinsonism effect of Hytrin that regulates energy homeostasis via activation of mitochondrial redox regulator; IDH2 leading to attenuation of synucleinopathy. However, the burst release kinetics of Hytrin restricts its therapeutic potential. Therefore, we aimed to improve Hytrin release kinetics through nanocarrier mediated delivery, replenish dopamine and serotonin by formulating Hytrin loaded polydopamine serotonin nanohybrid for PD protection. Present study also explores IDH2 mediated neuroprotective action in retardation of synucleinopathy for PD prevention. Nanoformulation has shown effective neurotherapeutic potential by improving Hytrin release profile in the reduction of PD symptoms in vitro and ex vivo. The neuroprotective effect has been attributed to IDH2 induction and alpha-synuclein reduction against rotenone insults. The direct physical interaction of IDH2 and alpha-synuclein, PD hallmark has been uncovered. The study divulges that the restorative effect of our nanoformulation significantly retards the PD deficits byinducing IDH2 mediated alpha-synuclein ubiquitination and proteasomal degradation pathway.
Collapse
Affiliation(s)
- Mohammed Nadim Sardoiwala
- Institute of Nano Science and Technology, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India
| | - Soni Jignesh Mohanbhai
- Institute of Nano Science and Technology, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India
| | - Surajit Karmakar
- Institute of Nano Science and Technology, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India.
| | - Subhasree Roy Choudhury
- Institute of Nano Science and Technology, Knowledge City, Sector-81, SAS Nagar, Mohali 140306, Punjab, India.
| |
Collapse
|
6
|
Mechanical Stretching-Induced Traumatic Brain Injury Is Mediated by the Formation of GSK-3β-Tau Complex to Impair Insulin Signaling Transduction. Biomedicines 2021; 9:biomedicines9111650. [PMID: 34829879 PMCID: PMC8615493 DOI: 10.3390/biomedicines9111650] [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: 09/16/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury confers a significant and growing public health burden. It is a major environmental risk factor for dementia. Nonetheless, the mechanism by which primary mechanical injury leads to neurodegeneration and an increased risk of dementia-related diseases is unclear. Thus, we aimed to investigate the effect of stretching on SH-SY5Y neuroblastoma cells that proliferate in vitro. These cells retain the dopamine-β-hydroxylase activity, thus being suitable for neuromechanistic studies. SH-SY5Y cells were cultured on stretchable membranes. The culture conditions contained two groups, namely non-stretched (control) and stretched. They were subjected to cyclic stretching (6 and 24 h) and 25% elongation at 1 Hz. Following stretching at 25% and 1 Hz for 6 h, the mechanical injury changed the mitochondrial membrane potential and triggered oxidative DNA damage at 24 h. Stretching decreased the level of brain-derived neurotrophic factors and increased amyloid-β, thus indicating neuronal stress. Moreover, the mechanical injury downregulated the insulin pathway and upregulated glycogen synthase kinase 3β (GSK-3β)S9/p-Tau protein levels, which caused a neuronal injury. Following 6 and 24 h of stretching, GSK-3βS9 was directly bound to p-TauS396. In contrast, the neuronal injury was improved using GSK-3β inhibitor TWS119, which downregulated amyloid-β/p-Taus396 phosphorylation by enhancing ERK1/2T202/Y204 and AktS473 phosphorylation. Our findings imply that the neurons were under stress and that the inactivation of the GSK3β could alleviate this defect.
Collapse
|
7
|
Shoemaker AR, Jones IE, Jeffris KD, Gabrielli G, Togliatti AG, Pichika R, Martin E, Kiskinis E, Franz CK, Finan J. Biofidelic dynamic compression of human cortical spheroids reproduces neurotrauma phenotypes. Dis Model Mech 2021; 14:273823. [PMID: 34746950 PMCID: PMC8713991 DOI: 10.1242/dmm.048916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 11/02/2021] [Indexed: 11/20/2022] Open
Abstract
Fundamental questions about patient heterogeneity and human-specific pathophysiology currently obstruct progress towards a therapy for traumatic brain injury (TBI). Human in vitro models have the potential to address these questions. 3D spheroidal cell culture protocols for human-origin neural cells have several important advantages over their 2D monolayer counterparts. Three dimensional spheroidal cultures may mature more quickly, develop more biofidelic electrophysiological activity and/or reproduce some aspects of brain architecture. Here, we present the first human in vitro model of non-penetrating TBI employing 3D spheroidal cultures. We used a custom-built device to traumatize these spheroids in a quantifiable, repeatable and biofidelic manner and correlated the heterogeneous, mechanical strain field with the injury phenotype. Trauma reduced cell viability, mitochondrial membrane potential and spontaneous, synchronous, electrophysiological activity in the spheroids. Electrophysiological deficits emerged at lower injury severities than changes in cell viability. Also, traumatized spheroids secreted lactate dehydrogenase, a marker of cell damage, and neurofilament light chain, a promising clinical biomarker of neurotrauma. These results demonstrate that 3D human in vitro models can reproduce important phenotypes of neurotrauma in vitro.
Collapse
Affiliation(s)
- Aaron R Shoemaker
- Department of Neurosurgery, NorthShore University Health System, Evanston, IL, USA
| | - Ian E Jones
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Kira D Jeffris
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Gina Gabrielli
- Department of Neurosurgery, NorthShore University Health System, Evanston, IL, USA
| | | | - Rajeswari Pichika
- Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eric Martin
- Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Colin K Franz
- Shirley Ryan AbilityLab, Chicago, IL, USA.,Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - John Finan
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
8
|
Wu YH, Rosset S, Lee TR, Dragunow M, Park T, Shim V. In Vitro Models of Traumatic Brain Injury: A Systematic Review. J Neurotrauma 2021; 38:2336-2372. [PMID: 33563092 DOI: 10.1089/neu.2020.7402] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health challenge that is also the third leading cause of death worldwide. It is also the leading cause of long-term disability in children and young adults worldwide. Despite a large body of research using predominantly in vivo and in vitro rodent models of brain injury, there is no medication that can reduce brain damage or promote brain repair mainly due to our lack of understanding in the mechanisms and pathophysiology of the TBI. The aim of this review is to examine in vitro TBI studies conducted from 2008-2018 to better understand the TBI in vitro model available in the literature. Specifically, our focus was to perform a detailed analysis of the in vitro experimental protocols used and their subsequent biological findings. Our review showed that the uniaxial stretch is the most frequently used way of load application, accounting for more than two-thirds of the studies reviewed. The rate and magnitude of the loading were varied significantly from study to study but can generally be categorized into mild, moderate, and severe injuries. The in vitro studies reviewed here examined key processes in TBI pathophysiology such as membrane disruptions leading to ionic dysregulation, inflammation, and the subsequent damages to the microtubules and axons, as well as cell death. Overall, the studies examined in this review contributed to the betterment of our understanding of TBI as a disease process. Yet, our review also revealed the areas where more work needs to be done such as: 1) diversification of load application methods that will include complex loading that mimics in vivo head impacts; 2) more widespread use of human brain cells, especially patient-matched human cells in the experimental set-up; and 3) need for building a more high-throughput system to be able to discover effective therapeutic targets for TBI.
Collapse
Affiliation(s)
- Yi-Han Wu
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Samuel Rosset
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Tae-Rin Lee
- Advanced Institute of Convergence Technology, Seoul National University, Seoul, Korea
| | - Mike Dragunow
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Thomas Park
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Vickie Shim
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
9
|
Lee IJ, Chao CY, Yang YC, Cheng JJ, Huang CL, Chiou CT, Huang HT, Kuo YH, Huang NK. Huang Lian Jie Du Tang attenuates paraquat-induced mitophagy in human SH-SY5Y cells: A traditional decoction with a novel therapeutic potential in treating Parkinson's disease. Biomed Pharmacother 2020; 134:111170. [PMID: 33383311 DOI: 10.1016/j.biopha.2020.111170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 10/22/2022] Open
Abstract
Huang Lian Jie Du Tang (HLJDT) is a traditional Chinese medical decoction for heat-fire clearing and detoxication. Theoretically, the cause of Parkinson's disease (PD) has been attributed to the dysregulations of internal wind, phlegm, fire, and stasis. Thus, HLJDT has been used to treat PD. However, the molecular mechanism is unknown. Besides, paraquat (PQ) as an herbicide has been known to impair midbrain dopaminergic neurons, resemblance to the pathology of PD. Thus, the molecular mechanism of HLJDT in treating PD and PQ-induced in vitro PD model was investigated in this study. Primarily, the dose-response of PQ (0.1∼1 mM)-induced neurotoxicity for 24 h was performed in the human neuroblastoma SH-SY5Y cells. The LD50 of PQ is around 0.3 mM and was applied throughout the following experiments. The neutral red assay was used to estimate cell viability. Co-transfection of the mitochondrial marker and proapoptotic factor genes were applied to measure the release of mitochondrial proapoptotic factors during PQ intoxication and HLJDT protection. The fluorescent dyes were used to detect mitochondrial membrane potential and free radical formation. Western blot and dot-blot analysis and immunocytochemistry were used to estimate the level of proteins related to apoptosis and mitophagy. PINK1 gene silencing was used to determine the significance of mitophagy during PQ intoxication. In this study, HLJDT attenuated PQ-induced apoptosis in SH-SY5Y cells. HLJDT reversed PQ-induced decreased mitochondrial membrane potential and suppressed PQ-induced increased cytosolic and mitochondrial free radical formations and mitochondrial proapoptotic factor releases. Furthermore, HLJDT mitigated PQ-induced increases in full-length PINK1, phosphorylations of Parkin and ubiquitin, mitochondrial translocation of phosphorylated Parkin, and mitophagy. PINK1 gene silencing attenuated PQ-induced neurotoxicity. Therefore, HLJDT attenuated PQ-induced cell death by regulating mitophagy.
Collapse
Affiliation(s)
- I-Jung Lee
- Herbal Medicine Department, Yokohama University of Pharmacy, Yokohama, Japan
| | - Che-Yi Chao
- Department of Psychiatry, Cardinal Tien Hospital, New Taipei City 23148, Taiwan, ROC
| | - Ying-Chen Yang
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 26047, Taiwan, ROC
| | - Jing-Jy Cheng
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11267, Taiwan, ROC
| | - Chuen-Lin Huang
- Medical Research Center, Cardinal Tien Hospital, Hsintien, New Taipei City 23148, Taiwan, ROC; Graduate Institute of Physiology & Department of Physiology and Biophysics, National Defense Medical Center, Taipei 11490, Taiwan, ROC
| | - Chun-Tang Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11267, Taiwan, ROC
| | - Hung-Tse Huang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11267, Taiwan, ROC
| | - Yao-Haur Kuo
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11267, Taiwan, ROC; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan, ROC
| | - Nai-Kuei Huang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 11267, Taiwan, ROC; The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan, ROC; Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei 11031, Taiwan, ROC.
| |
Collapse
|
10
|
Sardoiwala MN, Karmakar S, Choudhury SR. Chitosan nanocarrier for FTY720 enhanced delivery retards Parkinson's disease via PP2A-EzH2 signaling in vitro and ex vivo. Carbohydr Polym 2020; 254:117435. [PMID: 33357908 DOI: 10.1016/j.carbpol.2020.117435] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/05/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) develops due to oxidative stress, mitochondrial aberrations, posttranslational modification, and α-Synuclein (α-Syn) aggregation. The α-synucleinopathy is attributed to phosphorylation and aggregation of α-Syn. A strategy to degrade or reduce phosphorylated protein paves the way to develop PD therapy. Hence, the neuroprotective efficiency of PP2A (Protein phosphatase 2) activator FTY720, loaded chitosan nanoformulation has been evaluated in vitro and ex vivo experimental PD models. Bio-compatible chitosan-based nanocarriers have been utilized to enhance the bio-availability and neuroprotective effect of FTY720. The neuroprotective effect of characterized nanoformulation was determined by the downregulation of PD hallmark phospho-serine 129 (pSer129) α-Syn, with anti-oxidative and anti-inflammatory potentials. The neuroprotective mechanism uncovered novel physical interaction of PP2A and polycomb group of protein Enhancer of zeste homolog 2 to mediate ubiquitination and degradation of agglomerated pSer129 α-Syn. Indeed, this study establishes the neuroprotective potential of chitosan based FTY720 nanoformulations by PP2A mediated epigenetic regulation for PD prevention.
Collapse
Affiliation(s)
| | - Surajit Karmakar
- Institute of Nano Science and Technology, Habitat Centre, Phase-10, Mohali, Punjab, 160062, India
| | - Subhasree Roy Choudhury
- Institute of Nano Science and Technology, Habitat Centre, Phase-10, Mohali, Punjab, 160062, India.
| |
Collapse
|
11
|
Raymont V, Thayanandan T. What do we know about the risks of developing dementia after traumatic brain injury? Minerva Med 2020; 112:288-297. [PMID: 33164474 DOI: 10.23736/s0026-4806.20.07084-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Traumatic brain injury (TBI) is a risk factor for the later development of dementia, but although the evidence dates back to the early 20th century, the nature of any association and its mechanistic pathways remain unclear. There has been greater focus on this subject over recent years, in part because of increasing reports around sports related TBIs, especially in the USA. Differences in research methods and clinical sampling remain the primary reason for the variable findings, although there is clearly increased prevalence of neurodegenerative disorders in general. Duration of follow up, definition of both TBI and dementia, and differences in the extent to which other dementia risk factors are controlled, as well as concerns about medical record accuracy are all issues yet to be resolved in TBI research, as is an absence pathological evidence. In addition, TBI has been reported to initiate a cascade of pathological processes related to several neurodegenerative disorders, and as such, it is likely that the risks vary between individuals. Given the evidence that dementia risk may increase with injury severity and frequency, a detailed account of age and type of injury, as well as lifetime TBI exposure is essential to document in future studies, and further longitudinal research with biomarker assessments are needed.
Collapse
Affiliation(s)
- Vanessa Raymont
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK -
| | - Tony Thayanandan
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
Yi L, Juan W, Gang C, Leiming Z, Jianning Z. Seawater Immersion Aggravates Early Mitochondrial Dysfunction and Increases Neuronal Apoptosis After Traumatic Brain Injury. Cell Mol Neurobiol 2020; 40:447-457. [PMID: 31667702 DOI: 10.1007/s10571-019-00747-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability in naval warfare. Due to the unique physiochemical properties of seawater, immersion in it exacerbates TBI and induces severe neural damage and complications. However, the characteristics and underlying mechanisms of seawater-immersed TBI remain unclear. Mitochondrial dysfunction is a major cause of TBI-associated brain damage because it leads to oxidative stress, decrease in energy production, and apoptosis. Thus, the present study aimed to further elucidate the current understanding of the pathology of seawater-immersed TBI, particularly the role of mitochondrial dysfunction, using a well-defined rat model of fluid percussion injury and a stretch injury model comprising cultured neurons. The biochemical and pathological markers of brain-related and neuronal injuries were evaluated. Histological analysis suggested that seawater immersion enhanced brain tissue injury and induced a significant increase in apoptosis in rats with TBI. Additionally, lactate dehydrogenase release occurred earlier and at higher levels in stretched neurons at 24 h after seawater immersion, which was consistent with more severe morphological changes and enhanced apoptosis. Furthermore, seawater immersion induced more rapid decreases in mitochondrial membrane potential, adenosine triphosphate (ATP) content, and H+-ATPase activity in the cortices of TBI rats. In addition, the immunochemical results revealed that seawater immersion further attenuated mitochondrial function in neurons exposed to stretch injury. The increases in neuronal damage and apoptosis triggered by seawater immersion were positively correlated with mitochondrial dysfunction in both in vivo and in vitro models. Thus, the present findings strengthen the current understanding of seawater-immersed TBI. Moreover, because seawater immersion aggravates mitochondrial dysfunction and contributes to post-traumatic neuronal cell death, it is important to consider mitochondria as a therapeutic target for seawater-immersed TBI.
Collapse
Affiliation(s)
- Liu Yi
- Department of Neurosurgery, General Navy Hospital of PLA, The Second Military Medical University, 6 Fucheng Road, Beijing, 100048, China
| | - Wang Juan
- Department of Neurosurgery, General Navy Hospital of PLA, The Second Military Medical University, 6 Fucheng Road, Beijing, 100048, China
| | - Cheng Gang
- Department of Neurosurgery, General Navy Hospital of PLA, The Second Military Medical University, 6 Fucheng Road, Beijing, 100048, China
| | - Zhang Leiming
- Department of Neurosurgery, General Navy Hospital of PLA, The Second Military Medical University, 6 Fucheng Road, Beijing, 100048, China
| | - Zhang Jianning
- Department of Neurosurgery, General Navy Hospital of PLA, The Second Military Medical University, 6 Fucheng Road, Beijing, 100048, China.
| |
Collapse
|
13
|
Rosas-Hernandez H, Burks SM, Cuevas E, Ali SF. Stretch-Induced Deformation as a Model to Study Dopaminergic Dysfunction in Traumatic Brain Injury. Neurochem Res 2019; 44:2546-2555. [DOI: 10.1007/s11064-019-02872-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 01/08/2023]
|
14
|
Fernandes J, Chandler JD, Liu KH, Uppal K, Hao L, Hu X, Go YM, Jones DP. Metabolomic Responses to Manganese Dose in SH-SY5Y Human Neuroblastoma Cells. Toxicol Sci 2019; 169:84-94. [PMID: 30715528 PMCID: PMC6484887 DOI: 10.1093/toxsci/kfz028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Manganese (Mn)-associated neurotoxicity has been well recognized. However, Mn is also an essential nutrient to maintain physiological function. Our previous study of human neuroblastoma SH-SY5Y cells showed that Mn treatment comparable to physiological and toxicological concentrations in human brain resulted in different mitochondrial responses, yet cellular metabolic responses associated with such different outcomes remain uncharacterized. Herein, SH-SY5Y cells were examined for metabolic responses discriminated by physiological and toxicological levels of Mn using high-resolution metabolomics (HRM). Before performing HRM, we examined Mn dose (from 0 to100 μM) and time effects on cell death. Although we did not observe any immediate cell death after 5 h exposure to any of the Mn concentrations assessed (0-100 μM), cell loss was present after a 24-h recovery period in cultures treated with Mn ≥ 50 μM. Exposure to Mn for 5 h resulted in a wide range of changes in cellular metabolism including amino acids (AA), neurotransmitters, energy, and fatty acids metabolism. Adaptive responses at 10 μM showed increases in neuroprotective AA metabolites (creatine, phosphocreatine, phosphoserine). A 5-h exposure to 100 µM Mn, a time before any cell death occurred, resulted in decreases in energy and fatty acid metabolites (hexose-1,6 bisphosphate, acyl carnitines). The results show that adjustments in AA metabolism occur in response to Mn that does not cause cell death while disruption in energy and fatty acid metabolism occur in response to Mn that results in subsequent cell death. The present study establishes utility for metabolomics analyses to discriminate adaptive and toxic molecular responses in a human in vitro cellular model that could be exploited in evaluation of Mn toxicity.
Collapse
Affiliation(s)
- Jolyn Fernandes
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Joshua D Chandler
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Ken H Liu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Karan Uppal
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Li Hao
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Xin Hu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
15
|
Liang X, Wang Z, Gao M, Wu S, Zhang J, Liu Q, Yu Y, Wang J, Liu W. Cyclic stretch induced oxidative stress by mitochondrial and NADPH oxidase in retinal pigment epithelial cells. BMC Ophthalmol 2019; 19:79. [PMID: 30885167 PMCID: PMC6421648 DOI: 10.1186/s12886-019-1087-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/12/2019] [Indexed: 01/24/2023] Open
Abstract
Background Vitreomacular adhesion (VMA) has been reported to associated with age-related macular degeneration (AMD). Understanding the mechanisms underlying cyclic stretch induced in retinal pigment epithelial cells (RPE) may be important for the treatment of VMA-related AMD. Method Cyclic stretch (1HZ, 20% elongation) was applied to cultured ARPE-19 cells for 15 min, 2 h, 6 h, 12 h, 24 h by flexcell FX-5000 Tension system. Total reactive oxygen species (ROS) were detected using DCFH-DA. Mitochondrial superoxide were detected using MitoSOX Red mitochondrial superoxide indicator. NADPH oxidases (NOX) and signaling pathways, such as p38 and PKC, were detected using western blot. Apocycin (Apo) were used as NOX inhibitors. Result High levels of total ROS were detected from 15 min to 24 h, whereas mitochondrial superoxide were higher only in early time. NOX2 were significantly increased at 24 h. NOX4 were significantly increased at 2 h and reach its peak at 24 h. P-p38 was significantly increased at 12 h and 24 h. P-PKC was significantly increased at 15 min and kept a persistent high level. The upregulated expression of NOX4 by cyclic stretch can be significantly decreased under p-PKC inhibitor other than p-p38 inhibitor. Conclusion Cyclic stretch induce oxidative stress from both mitochodrial and NADPH oxidase in RPE cells, which may prompt oxidative damage in VMA-related AMD.
Collapse
Affiliation(s)
- Xida Liang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Zengyi Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Meng Gao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Shen Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Jingxue Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Qian Liu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Yanping Yu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Jing Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Wu Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China.
| |
Collapse
|
16
|
Szarka N, Pabbidi MR, Amrein K, Czeiter E, Berta G, Pohoczky K, Helyes Z, Ungvari Z, Koller A, Buki A, Toth P. Traumatic Brain Injury Impairs Myogenic Constriction of Cerebral Arteries: Role of Mitochondria-Derived H 2O 2 and TRPV4-Dependent Activation of BK ca Channels. J Neurotrauma 2018; 35:930-939. [PMID: 29179622 DOI: 10.1089/neu.2017.5056] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) impairs autoregulation of cerebral blood flow, which contributes to the development of secondary brain injury, increasing mortality of patients. Impairment of pressure-induced myogenic constriction of cerebral arteries plays a critical role in autoregulatory dysfunction; however, the underlying cellular and molecular mechanisms are not well understood. To determine the role of mitochondria-derived H2O2 and large-conductance calcium-activated potassium channels (BKCa) in myogenic autoregulatory dysfunction, middle cerebral arteries (MCAs) were isolated from rats with severe weight drop-impact acceleration brain injury. We found that 24 h post-TBI MCAs exhibited impaired myogenic constriction, which was restored by treatment with a mitochondria-targeted antioxidant (mitoTEMPO), by scavenging of H2O2 (polyethylene glycol [PEG]-catalase) and by blocking both BKCa channels (paxilline) and transient receptor potential cation channel subfamily V member 4 (TRPV4) channels (HC 067047). Further, exogenous administration of H2O2 elicited significant dilation of MCAs, which was inhibited by blocking either BKCa or TRPV4 channels. Vasodilation induced by the TRPV4 agonist GSK1016790A was inhibited by paxilline. In cultured vascular smooth muscle cells H2O2 activated BKCa currents, which were inhibited by blockade of TRPV4 channels. Collectively, our results suggest that after TBI, excessive mitochondria-derived H2O2 activates BKCa channels via a TRPV4-dependent pathway in the vascular smooth muscle cells, which impairs pressure-induced constriction of cerebral arteries. Future studies should elucidate the therapeutic potential of pharmacological targeting of this pathway in TBI, to restore autoregulatory function in order to prevent secondary brain damage and decrease mortality.
Collapse
Affiliation(s)
- Nikolett Szarka
- Cerebrovascular Laboratory, Department of Neurosurgery, Medical School University of Pecs, Pecs. Hungary.,Neurotrauma Research Group, Janos Szentagothai Research Center, Medical School University of Pecs, Pecs. Hungary.,Department of Translational Medicine, Medical School University of Pecs, Pecs. Hungary
| | - Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Krisztina Amrein
- Cerebrovascular Laboratory, Department of Neurosurgery, Medical School University of Pecs, Pecs. Hungary.,Neurotrauma Research Group, Janos Szentagothai Research Center, Medical School University of Pecs, Pecs. Hungary
| | - Endre Czeiter
- Cerebrovascular Laboratory, Department of Neurosurgery, Medical School University of Pecs, Pecs. Hungary.,Neurotrauma Research Group, Janos Szentagothai Research Center, Medical School University of Pecs, Pecs. Hungary.,MTA-PTE Clinical Neuroscience MR Research Group, Pecs, Hungary
| | - Gergely Berta
- Department of Medical Biology, Medical School University of Pecs, Pecs. Hungary
| | - Krisztina Pohoczky
- Department of Pharmacology and Pharmacotherapy, Medical School University of Pecs, Pecs. Hungary.,MTA-PTE NAP B Chronic Pain Research Group, Pecs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School University of Pecs, Pecs. Hungary.,MTA-PTE NAP B Chronic Pain Research Group, Pecs, Hungary
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Akos Koller
- Cerebrovascular Laboratory, Department of Neurosurgery, Medical School University of Pecs, Pecs. Hungary.,Institute of Natural Sciences, University of Physical Education, Budapest, Hungary.,Department of Physiology, New York Medical College, Valhalla, New York
| | - Andras Buki
- Cerebrovascular Laboratory, Department of Neurosurgery, Medical School University of Pecs, Pecs. Hungary.,Neurotrauma Research Group, Janos Szentagothai Research Center, Medical School University of Pecs, Pecs. Hungary
| | - Peter Toth
- Cerebrovascular Laboratory, Department of Neurosurgery, Medical School University of Pecs, Pecs. Hungary.,Neurotrauma Research Group, Janos Szentagothai Research Center, Medical School University of Pecs, Pecs. Hungary.,Department of Translational Medicine, Medical School University of Pecs, Pecs. Hungary.,MTA-PTE Clinical Neuroscience MR Research Group, Pecs, Hungary.,Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
17
|
Xu Z, Liu Y, Yang D, Yuan F, Ding J, Chen H, Tian H. Sesamin protects SH-SY5Y cells against mechanical stretch injury and promoting cell survival. BMC Neurosci 2017; 18:57. [PMID: 28784087 PMCID: PMC5547510 DOI: 10.1186/s12868-017-0378-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 08/01/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Sesamin is a well-known antioxidant extracted from sesame seeds that exhibits various curative effects. The present study investigated whether sesamin would protect neuroblastoma SH-SY5Y cells against mechanical stretch injury-induced increases in reactive oxygen species (ROS) and apoptosis. Additionally, the mechanisms underlying these actives were investigated. Following exposure to mechanical stretch injury, cells were incubated for further investigations. Lactate dehydrogenase and Cell Counting Kit-8 assays were used to assess cell viability, and a terminal deoxynucleotidyl transferase dUTP nick end labeling assay and flow cytometric analysis were performed to evaluate changes in mitochondrial membrane potential (ΔΨm). Furthermore, intracellular levels of ROS production were measured by 20, 70-dichlorofluorescein diacetate staining, the mRNA levels of matrix metallopeptidase 9 (MMP-9) were evaluated using real-time polymerase chain reaction analysis, and the determinations had also been made on related proteins by Western blot analysis. RESULTS Exposure to mechanical stretch injury significantly decreased cell viability but this decrease was attenuated by pretreatment with sesamin (50 μM). Sesamin also significantly inhibited mechanical stretch injury-induced increases in intracellular ROS production, attenuated declines in ΔΨm, diminished the expressions of pro-apoptotic proteins, and decreased cell apoptosis. Stretch injury increased Bax and cleaved caspase 3 levels, enhanced the gene expression of MMP-9, increased the phosphorylation levels of Akt, p38, and JNK and decreased Bcl-2 levels in the cells. However, pretreatment with sesamin reduced the mechanical stretch injury-induced overexpression of MMP-9. CONCLUSIONS Sesamin protected SH-SY5Y cells against stretch injury by attenuating increases in ROS levels and suppressing apoptosis. Accordingly, sesamin seems to be a potentially therapeutic agent in the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Zhiming Xu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO. 600, Yi Shan Road, Xuhui District, Shanghai, 200030, China
| | - Yingliang Liu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO. 600, Yi Shan Road, Xuhui District, Shanghai, 200030, China
| | - Dianxu Yang
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO. 600, Yi Shan Road, Xuhui District, Shanghai, 200030, China
| | - Fang Yuan
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO. 600, Yi Shan Road, Xuhui District, Shanghai, 200030, China
| | - Jun Ding
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO. 600, Yi Shan Road, Xuhui District, Shanghai, 200030, China
| | - Hao Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO. 600, Yi Shan Road, Xuhui District, Shanghai, 200030, China
| | - Hengli Tian
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO. 600, Yi Shan Road, Xuhui District, Shanghai, 200030, China.
| |
Collapse
|
18
|
Li F, Ge B, Damirin A. Overexpression of p58ipk protects neuroblastoma against paraquat-induced toxicity. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8233-8242. [PMID: 31966674 PMCID: PMC6965456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/20/2017] [Indexed: 06/10/2023]
Abstract
BACKGROUND Paraquat (PQ) is a powerful pathologic pesticide that contribute to the neurotoxicity, however, the pathogenic mechanism between them was unclear. The aims of this study were to explore the underlying mechanism of PQ-induced toxicity and then make potential contribute to such neuronal diseases therapy. METHODS Human cell line SH-SY5Y was pretreated with a set concentrations of PQ to detect the cell apoptosis and the expression of related genes and proteins. Next, pcDNA 3.1-p58ipk or si-p58ipk was transfected the PQ-induced cells to detect the cytotoxicity. RESULTS PQ significantly increased the cell apoptosis as well as the expression of p58ipk and CHOP, but decreased the expression of pAKT. p58ipk suppression resulted in an increase of cell apoptosis and CHOP expression, but the expression of pAKT was significantly decreased in PQ-induced SH-SY5Y cells. However, overexpressed p58ipk led to an opposite result. CONCLUSION The results indicated that the expression of p58ipk was related to the toxicity level of PQ-induced cells and the mechanism between them was that p58ipk regulated the toxicity might through regulating the endoplasmic reticulum stress (ER-stress) and then regulating cell apoptosis. Further studies take emphasize on the effect of ER-stress on neuron system and explore ER-stress-related therapy are important on the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Fengrui Li
- School of Life Sciences, Inner Mongolia UniversityHohhot 010021, China
- Department of Forensic Medicine, Baotou Medical CollegeBaotou, China
| | - Biao Ge
- Department of Geriatrics, Baogang Hospital of Inner MongoliaBaotou, China
| | | |
Collapse
|
19
|
Hichor M, Sampathkumar NK, Montanaro J, Borderie D, Petit PX, Gorgievski V, Tzavara ET, Eid AA, Charbonnier F, Grenier J, Massaad C. Paraquat Induces Peripheral Myelin Disruption and Locomotor Defects: Crosstalk with LXR and Wnt Pathways. Antioxid Redox Signal 2017; 27:168-183. [PMID: 27788593 DOI: 10.1089/ars.2016.6711] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AIMS Paraquat (PQT), a redox-active herbicide, is a free radical-producing molecule, causing damage particularly to the nervous system; thus, it is employed as an animal model for Parkinson's disease. However, its impact on peripheral nerve demyelination is still unknown. Our aim is to decipher the influence of PQT-induced reactive oxygen species (ROS) production on peripheral myelin. RESULTS We report that PQT provokes severe locomotor and sensory defects in mice. PQT elicited an oxidative stress in the nerve, resulting in an increase of lipid peroxidation and protein carbonylation, despite the induction of nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent antioxidant defenses. We observed a dramatic disorganization of myelin sheaths in the sciatic nerves, dysregulation of myelin gene expression, and aggregation of myelin proteins, a hallmark of demyelination. PQT altered myelin gene expression via liver X receptor (LXR) signaling, a negative regulator of peripheral myelin gene expression through its dialog with the Wnt/β-catenin pathway. PQT prevented β-catenin binding on myelin gene promoters, resulting in the inhibition of Wnt/β-catenin-dependent myelin gene expression. Wnt pathway activation by LiCl dampened the deleterious effects of PQT. LiCl blocked PQT-induced oxidative stress and reduced Schwann cell death. LiCl+PQT-treated mice had normal sensorimotor behaviors and a usual nerve structure. INNOVATION We reveal that PQT damages the sciatic nerve by generating an oxidative stress, dysregulating LXR and Wnt/β-catenin pathways. The activation of Wnt signaling by LiCl reduced the deleterious effects of PQT on the nerve. CONCLUSION We demonstrate that PQT instigates peripheral nerve demyelinating neuropathies by enhancing ROS production and deregulating LXR and Wnt pathways. Stimulating Wnt pathway could be a therapeutic strategy for neuropathy treatment. Antioxid. Redox Signal. 27, 168-183.
Collapse
Affiliation(s)
- Mehdi Hichor
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Nirmal Kumar Sampathkumar
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Julia Montanaro
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Didier Borderie
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Patrice X Petit
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Victor Gorgievski
- 2 INSERM UMRS-S 1130, CNRS UMR824, Pierre and Marie Curie University , Paris, France
| | - Eleni T Tzavara
- 2 INSERM UMRS-S 1130, CNRS UMR824, Pierre and Marie Curie University , Paris, France
| | - Assaad A Eid
- 3 Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Frédéric Charbonnier
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Julien Grenier
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Charbel Massaad
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| |
Collapse
|
20
|
Paraquat and maneb co-exposure induces noradrenergic locus coeruleus neurodegeneration through NADPH oxidase-mediated microglial activation. Toxicology 2017; 380:1-10. [DOI: 10.1016/j.tox.2017.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/24/2017] [Accepted: 02/09/2017] [Indexed: 01/31/2023]
|
21
|
Jia Y, Yuan H, Shan S, Xu G, Yu J, Zhao C, Mou X. Corosolic acid inhibits the proliferation of osteosarcoma cells by inducing apoptosis. Oncol Lett 2016; 12:4187-4194. [PMID: 27895790 DOI: 10.3892/ol.2016.5185] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/25/2016] [Indexed: 11/05/2022] Open
Abstract
Corosolic acid (CRA), a pentacyclic triterpene isolated from medicinal herbs, has been reported to exhibit anticancer properties in several cancers. However, the anticancer activity of CRA in osteosarcoma cells is still unclear. In the present study, the inhibitory effect of CRA in osteosarcoma MG-63 cells was investigated, and the results revealed that CRA significantly inhibited the viability of MG-63 cells in a dose- and time-dependent manner. A typical apoptotic hallmark such as DNA ladder was detected by agarose gel electrophoresis following treatment with CRA. Further experiments demonstrated that CRA induced apoptosis of MG-63 cells by flow cytometry using propidium iodide and annexin V staining. In addition, it was observed that the apoptosis of MG-63 cells induced by CRA was closely associated with activation of caspase-3 and caspase-9, loss of mitochondrial membrane potential, and release of cytochrome c from mitochondria, suggesting that CRA may trigger the activation of the mitochondria-mediated apoptosis pathway. In addition, the inhibition of caspase activity attenuated the CRA-induced apoptosis of MG-63 cells, which further confirmed the role of the mitochondrial pathway in CRA-induced apoptosis. These results indicated that CRA could induce the apoptosis of osteosarcoma cells through activating the mitochondrial pathway, which provides an evidence that CRA may be a useful chemotherapeutic agent for osteosarcoma.
Collapse
Affiliation(s)
- Yong Jia
- Orthopedic Center of Chinese PLA, Urumqi General Hospital of Lanzhou Military Region, Urumqi, Xinjiang 830000, P.R. China
| | - Hua Yuan
- Department of Rehabilitation, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Shouqin Shan
- Department of Rehabilitation, Qingdao First Sanatorium of Jinan Military Region, Qingdao, Shandong 266071, P.R. China
| | - Gang Xu
- Orthopedic Center of Chinese PLA, Urumqi General Hospital of Lanzhou Military Region, Urumqi, Xinjiang 830000, P.R. China
| | - Jie Yu
- Department of Information, Urumqi General Hospital of Lanzhou Military Region, Urumqi, Xinjiang 830000, P.R. China
| | - Chenguang Zhao
- Department of Rehabilitation, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiang Mou
- Department of Rehabilitation, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
22
|
Acetazolamide Mitigates Astrocyte Cellular Edema Following Mild Traumatic Brain Injury. Sci Rep 2016; 6:33330. [PMID: 27623738 PMCID: PMC5022024 DOI: 10.1038/srep33330] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 08/25/2016] [Indexed: 12/31/2022] Open
Abstract
Non-penetrating or mild traumatic brain injury (mTBI) is commonly experienced in accidents, the battlefield and in full-contact sports. Astrocyte cellular edema is one of the major factors that leads to high morbidity post-mTBI. Various studies have reported an upregulation of aquaporin-4 (AQP4), a water channel protein, following brain injury. AZA is an antiepileptic drug that has been shown to inhibit AQP4 expression and in this study we investigate the drug as a therapeutic to mitigate the extent of mTBI induced cellular edema. We hypothesized that mTBI-mediated astrocyte dysfunction, initiated by increased intracellular volume, could be reduced when treated with AZA. We tested our hypothesis in a three-dimensional in vitro astrocyte model of mTBI. Samples were subject to no stretch (control) or one high-speed stretch (mTBI) injury. AQP4 expression was significantly increased 24 hours after mTBI. mTBI resulted in a significant increase in the cell swelling within 30 min of mTBI, which was significantly reduced in the presence of AZA. Cell death and expression of S100B was significantly reduced when AZA was added shortly before mTBI stretch. Overall, our data point to occurrence of astrocyte swelling immediately following mTBI, and AZA as a promising treatment to mitigate downstream cellular mortality.
Collapse
|
23
|
Ganapathy K, Datta I, Sowmithra S, Joshi P, Bhonde R. Influence of 6-Hydroxydopamine Toxicity on α-Synuclein Phosphorylation, Resting Vesicle Expression, and Vesicular Dopamine Release. J Cell Biochem 2016; 117:2719-2736. [DOI: 10.1002/jcb.25570] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 04/07/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Kavina Ganapathy
- School of Regenerative Medicine; Manipal University; Bengaluru Karnataka India
| | - Indrani Datta
- Department of Biophysics; National Institute of Mental Health and Neurosciences, an Institute of National Importance; Bengaluru Karnataka India
| | - Sowmithra Sowmithra
- Department of Biophysics; National Institute of Mental Health and Neurosciences, an Institute of National Importance; Bengaluru Karnataka India
| | - Preeti Joshi
- Department of Biophysics; National Institute of Mental Health and Neurosciences, an Institute of National Importance; Bengaluru Karnataka India
| | - Ramesh Bhonde
- School of Regenerative Medicine; Manipal University; Bengaluru Karnataka India
| |
Collapse
|
24
|
Turner RC, Lucke-Wold BP, Robson MJ, Lee JM, Bailes JE. Alzheimer's disease and chronic traumatic encephalopathy: Distinct but possibly overlapping disease entities. Brain Inj 2016; 30:1279-1292. [PMID: 27715315 PMCID: PMC5303562 DOI: 10.1080/02699052.2016.1193631] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE) have long been recognized as sharing some similar neuropathological features, mainly the presence of neurofibrilary tangles and hyperphosphorylated tau, but have generally been described as distinct entities. Evidence indicates that neurotrauma increases the risk of developing dementia and accelerates the progression of disease. Findings are emerging that CTE and AD may be present in the same patients. CLINICAL PRESENTATION This study presents a series of previously unpublished cases, with one case demonstrating possible neurotrauma-related AD, one pure CTE, and an example of a case exhibiting features of both AD and CTE. The future significance of this work lies not only in the confirmation of AD-CTE co-existence, but, more importantly, ways of generating a hypothesis about the possibility that CTE may accelerate AD development. Understanding the relationship between neurotrauma and neurodegenerative disease will help elucidate how distinct disease entities can co-exist in the same patient. It will ultimately require the use of pre-clinical animal models and repeat injury paradigms to investigate clinically relevant injury mechanisms. These models should produce a CTE-like phenotype that must be both neuropathologically and behaviourally similar to human disease. CONCLUSION This case series and review of the literature presents a discussion of AD and CTE in the context of neurotrauma. It highlights recent work from repetitive neurotrauma models with an emphasis on those exhibiting a CTE-like phenotype. Potential mechanisms of interest shared amongst AD and CTE are briefly addressed and future experiments are advocated for to enhance understanding of CTE pathophysiology and the relationship between CTE and AD.
Collapse
Affiliation(s)
- Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV 26506
- Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Matthew J. Robson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John M. Lee
- NorthShore Neurological Institute, NorthShore University Health System, Evanston, IL 60201
| | - Julian E. Bailes
- NorthShore Neurological Institute, NorthShore University Health System, Evanston, IL 60201
| |
Collapse
|
25
|
Low-Dose Aronia melanocarpa Concentrate Attenuates Paraquat-Induced Neurotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:5296271. [PMID: 26770655 PMCID: PMC4684878 DOI: 10.1155/2016/5296271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/26/2015] [Accepted: 08/30/2015] [Indexed: 12/05/2022]
Abstract
Herbicides containing paraquat may contribute to the pathogenesis of neurodegenerative disorders such as Parkinson's disease. Paraquat induces reactive oxygen species-mediated apoptosis in neurons, which is a primary mechanism behind its toxicity. We sought to test the effectiveness of a commercially available polyphenol-rich Aronia melanocarpa (aronia berry) concentrate in the amelioration of paraquat-induced neurotoxicity. Considering the abundance of antioxidants in aronia berries, we hypothesized that aronia berry concentrate attenuates the paraquat-induced increase in reactive oxygen species and protects against paraquat-mediated neuronal cell death. Using a neuronal cell culture model, we observed that low doses of aronia berry concentrate protected against paraquat-mediated neurotoxicity. Additionally, low doses of the concentrate attenuated the paraquat-induced increase in superoxide, hydrogen peroxide, and oxidized glutathione levels. Interestingly, high doses of aronia berry concentrate increased neuronal superoxide levels independent of paraquat, while at the same time decreasing hydrogen peroxide. Moreover, high-dose aronia berry concentrate potentiated paraquat-induced superoxide production and neuronal cell death. In summary, aronia berry concentrate at low doses restores the homeostatic redox environment of neurons treated with paraquat, while high doses exacerbate the imbalance leading to further cell death. Our findings support that moderate levels of aronia berry concentrate may prevent reactive oxygen species-mediated neurotoxicity.
Collapse
|
26
|
Yin Y, Zhang J, Song D. Effects of lysine aspirin on lung AQP5 expression and lymphocyte apoptosis in paraquat-poisoned rats. TOXIN REV 2015. [DOI: 10.3109/15569543.2015.1015036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
27
|
Pahuja R, Seth K, Shukla A, Shukla RK, Bhatnagar P, Chauhan LKS, Saxena PN, Arun J, Chaudhari BP, Patel DK, Singh SP, Shukla R, Khanna VK, Kumar P, Chaturvedi RK, Gupta KC. Trans-blood brain barrier delivery of dopamine-loaded nanoparticles reverses functional deficits in parkinsonian rats. ACS NANO 2015; 9:4850-71. [PMID: 25825926 DOI: 10.1021/nn506408v] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Sustained and safe delivery of dopamine across the blood brain barrier (BBB) is a major hurdle for successful therapy in Parkinson's disease (PD), a neurodegenerative disorder. Therefore, in the present study we designed neurotransmitter dopamine-loaded PLGA nanoparticles (DA NPs) to deliver dopamine to the brain. These nanoparticles slowly and constantly released dopamine, showed reduced clearance of dopamine in plasma, reduced quinone adduct formation, and decreased dopamine autoxidation. DA NPs were internalized in dopaminergic SH-SY5Y cells and dopaminergic neurons in the substantia nigra and striatum, regions affected in PD. Treatment with DA NPs did not cause reduction in cell viability and morphological deterioration in SH-SY5Y, as compared to bulk dopamine-treated cells, which showed reduced viability. Herein, we report that these NPs were able to cross the BBB and capillary endothelium in the striatum and substantia nigra in a 6-hydroxydopamine (6-OHDA)-induced rat model of PD. Systemic intravenous administration of DA NPs caused significantly increased levels of dopamine and its metabolites and reduced dopamine-D2 receptor supersensitivity in the striatum of parkinsonian rats. Further, DA NPs significantly recovered neurobehavioral abnormalities in 6-OHDA-induced parkinsonian rats. Dopamine delivered through NPs did not cause additional generation of ROS, dopaminergic neuron degeneration, and ultrastructural changes in the striatum and substantia nigra as compared to 6-OHDA-lesioned rats. Interestingly, dopamine delivery through nanoformulation neither caused alterations in the heart rate and blood pressure nor showed any abrupt pathological change in the brain and other peripheral organs. These results suggest that NPs delivered dopamine into the brain, reduced dopamine autoxidation-mediated toxicity, and ultimately reversed neurochemical and neurobehavioral deficits in parkinsonian rats.
Collapse
Affiliation(s)
- Richa Pahuja
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
- §Academy of Scientific and Innovative Research (AcSIR), Delhi 110001, India
| | - Kavita Seth
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
| | - Anshi Shukla
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
| | - Rajendra Kumar Shukla
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
| | | | | | - Prem Narain Saxena
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
| | - Jharna Arun
- ∥CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226001, India
| | - Bhushan Pradosh Chaudhari
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
- §Academy of Scientific and Innovative Research (AcSIR), Delhi 110001, India
| | - Devendra Kumar Patel
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
- §Academy of Scientific and Innovative Research (AcSIR), Delhi 110001, India
| | - Sheelendra Pratap Singh
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
| | - Rakesh Shukla
- §Academy of Scientific and Innovative Research (AcSIR), Delhi 110001, India
- ∥CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow 226001, India
| | - Vinay Kumar Khanna
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
- §Academy of Scientific and Innovative Research (AcSIR), Delhi 110001, India
| | - Pradeep Kumar
- §Academy of Scientific and Innovative Research (AcSIR), Delhi 110001, India
| | - Rajnish Kumar Chaturvedi
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
- §Academy of Scientific and Innovative Research (AcSIR), Delhi 110001, India
| | - Kailash Chand Gupta
- †CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80 MG Marg, Lucknow 226001, India
- §Academy of Scientific and Innovative Research (AcSIR), Delhi 110001, India
| |
Collapse
|
28
|
Kochanek PM, Jackson TC, Ferguson NM, Carlson SW, Simon DW, Brockman EC, Ji J, Bayir H, Poloyac SM, Wagner AK, Kline AE, Empey PE, Clark RS, Jackson EK, Dixon CE. Emerging therapies in traumatic brain injury. Semin Neurol 2015; 35:83-100. [PMID: 25714870 PMCID: PMC4356170 DOI: 10.1055/s-0035-1544237] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite decades of basic and clinical research, treatments to improve outcomes after traumatic brain injury (TBI) are limited. However, based on the recent recognition of the prevalence of mild TBI, and its potential link to neurodegenerative disease, many new and exciting secondary injury mechanisms have been identified and several new therapies are being evaluated targeting both classic and novel paradigms. This includes a robust increase in both preclinical and clinical investigations. Using a mechanism-based approach the authors define the targets and emerging therapies for TBI. They address putative new therapies for TBI across both the spectrum of injury severity and the continuum of care, from the field to rehabilitation. They discussTBI therapy using 11 categories, namely, (1) excitotoxicity and neuronal death, (2) brain edema, (3) mitochondria and oxidative stress, (4) axonal injury, (5) inflammation, (6) ischemia and cerebral blood flow dysregulation, (7) cognitive enhancement, (8) augmentation of endogenous neuroprotection, (9) cellular therapies, (10) combination therapy, and (11) TBI resuscitation. The current golden age of TBI research represents a special opportunity for the development of breakthroughs in the field.
Collapse
Affiliation(s)
- Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Travis C. Jackson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nikki Miller Ferguson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shaun W. Carlson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departmentol Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dennis W. Simon
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Erik C. Brockman
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jing Ji
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hülya Bayir
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Samuel M. Poloyac
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Amy K. Wagner
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Anthony E. Kline
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Philip E. Empey
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Robert S.B. Clark
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Edwin K. Jackson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departmentol Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
29
|
Seo HJ, Choi SJ, Lee JH. Paraquat Induces Apoptosis through Cytochrome C Release and ERK Activation. Biomol Ther (Seoul) 2014; 22:503-9. [PMID: 25489417 PMCID: PMC4256029 DOI: 10.4062/biomolther.2014.115] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 10/25/2014] [Accepted: 11/04/2014] [Indexed: 01/18/2023] Open
Abstract
Paraquat has been suggested to induce apoptosis by generation of reactive oxygen species (ROS). However, little is known about the mechanism of paraquat-induced apoptosis. Here, we demonstrate that extracellular signal-regulated protein kinase (ERK) is required for paraquat-induced apoptosis in NIH3T3 cells. Paraquat treatment resulted in activation of ERK, and U0126, inhibitors of the MEK/ERK signaling pathway, prevented apoptosis. Moreover, paraquat-induced apoptosis was associated with cytochrome C release, which could be prevented by treatment with the MEK inhibitors. Taken together, our findings suggest that ERK activation plays an active role in mediating paraquat-induced apoptosis of NIH3T3 cells.
Collapse
Affiliation(s)
- Hong Joo Seo
- Department of Thoracic and Cardiovascular Surgery, Chosun University School of Medicine, Gwangju 501-759, Republic of Korea
| | - Sang Joon Choi
- Department of Obstetrics and Gynecology, Chosun University Hospital, Chosun University School of Medicine, Gwangju 501-759, Republic of Korea
| | - Jung-Hee Lee
- Department of Cellular and Molecular Medicine, Chosun University School of Medicine, Gwangju 501-759, Republic of Korea
| |
Collapse
|