1
|
Kalimon OJ, Vekaria HJ, Prajapati P, Short SL, Hubbard WB, Sullivan PG. The Uncoupling Effect of 17β-Estradiol Underlies the Resilience of Female-Derived Mitochondria to Damage after Experimental TBI. Life (Basel) 2024; 14:961. [PMID: 39202703 PMCID: PMC11355196 DOI: 10.3390/life14080961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Current literature finds females have improved outcomes over their male counterparts after severe traumatic brain injury (TBI), while the opposite seems to be true for mild TBI. This begs the question as to what may be driving these sex differences after TBI. Estrogen is thought to be neuroprotective in certain diseases, and its actions have been shown to influence mitochondrial function. Mitochondrial impairment is a major hallmark of TBI, and interestingly, this dysfunction has been shown to be more severe in males than females after brain injury. This suggests estrogen could be playing a role in promoting "mitoprotection" following TBI. Despite the existence of estrogen receptors in mitochondria, few studies have examined the direct role of estrogen on mitochondrial function, and no studies have explored this after TBI. We hypothesized ex vivo treatment of isolated mitochondria with 17β-estradiol (E2) would improve mitochondrial function after experimental TBI in mice. Total mitochondria from the ipsilateral (injured) and contralateral (control) cortices of male and female mice were isolated 24 h post-controlled severe cortical impact (CCI) and treated with vehicle, 2 nM E2, or 20 nM E2 immediately before measuring reactive oxygen species (ROS) production, bioenergetics, electron transport chain complex (ETC) activities, and β-oxidation of palmitoyl carnitine. Protein expression of oxidative phosphorylation (OXPHOS) complexes was also measured in these mitochondrial samples to determine whether this influenced functional outcomes with respect to sex or injury. While mitochondrial ROS production was affected by CCI in both sexes, there were other sex-specific patterns of mitochondrial injury 24 h following severe CCI. For instance, mitochondria from males were more susceptible to CCI-induced injury with respect to bioenergetics and ETC complex activities, whereas mitochondria from females showed only Complex II impairment and reduced β-oxidation after injury. Neither concentration of E2 influenced ETC complex activities themselves, but 20 nM E2 appeared to uncouple mitochondria isolated from the contralateral cortex in both sexes, as well as the injured ipsilateral cortex of females. These studies highlight the significance of measuring mitochondrial dysfunction in both sexes after TBI and also shed light on another potential neuroprotective mechanism in which E2 may attenuate mitochondrial dysfunction after TBI in vivo.
Collapse
Affiliation(s)
- Olivia J. Kalimon
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508, USA;
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA; (H.J.V.); (P.P.); (S.L.S.); (W.B.H.)
- Lexington Veterans Affairs Healthcare System, Lexington, KY 40502, USA
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA; (H.J.V.); (P.P.); (S.L.S.); (W.B.H.)
- Lexington Veterans Affairs Healthcare System, Lexington, KY 40502, USA
| | - Paresh Prajapati
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA; (H.J.V.); (P.P.); (S.L.S.); (W.B.H.)
- Lexington Veterans Affairs Healthcare System, Lexington, KY 40502, USA
| | - Sydney L. Short
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA; (H.J.V.); (P.P.); (S.L.S.); (W.B.H.)
| | - W. Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA; (H.J.V.); (P.P.); (S.L.S.); (W.B.H.)
- Lexington Veterans Affairs Healthcare System, Lexington, KY 40502, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA
| | - Patrick G. Sullivan
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508, USA;
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA; (H.J.V.); (P.P.); (S.L.S.); (W.B.H.)
- Lexington Veterans Affairs Healthcare System, Lexington, KY 40502, USA
| |
Collapse
|
2
|
Ziakova K, Kovalska M, Pilchova I, Dibdiakova K, Brodnanova M, Pokusa M, Kalenska D, Racay P. Involvement of Proteasomal and Endoplasmic Reticulum Stress in Neurodegeneration After Global Brain Ischemia. Mol Neurobiol 2023; 60:6316-6329. [PMID: 37452223 PMCID: PMC10533597 DOI: 10.1007/s12035-023-03479-5] [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: 01/10/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
A brief period of transient global brain ischemia leads to selective ischemic neurodegeneration associated with death of hippocampal CA1 pyramidal neurons days after reperfusion. The mechanism of such selective and delayed neurodegeneration is still uncertain. Our work aimed to study the involvement of proteasomal and endoplasmic reticulum (ER) stress in ischemic neurodegeneration. We have performed laser scanning confocal microscopy analysis of brain slices from control and experimental animals that underwent global brain ischemia for 15 min and varying times of reperfusion. We have focused on ubiquitin, PUMA, a proapoptotic protein of the Bcl-2 family overexpressed in response to both proteasomal and ER stress, and p53, which controls expression of PUMA. We have also examined the expression of HRD1, an E3 ubiquitin ligase that was shown to be overexpressed after ER stress. We have also examined potential crosstalk between proteasomal and ER stress using cellular models of both proteasomal and ER stress. We demonstrate that global brain ischemia is associated with an appearance of distinct immunoreactivity of ubiquitin, PUMA and p53 in pyramidal neurons of the CA1 layer of the hippocampus 72 h after ischemic insults. Such changes correlate with a delay and selectivity of ischemic neurodegeneration. Immunoreactivity of HRD1 observed in all investigated regions of rat brain was transiently absent in both CA1 and CA3 pyramidal neurones 24 h after ischemia in the hippocampus, which does not correlate with a delay and selectivity of ischemic neurodegeneration. We do not document significant crosstalk between proteasomal and ER stress. Our results favour dysfunction of the ubiquitin proteasome system and consequent p53-induced expression of PUMA as the main mechanisms responsible for selective and delayed degeneration of pyramidal neurons of the hippocampal CA1 layer in response to global brain ischemia.
Collapse
Affiliation(s)
- Katarina Ziakova
- Biomedical Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Ivana Pilchova
- Biomedical Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Katarina Dibdiakova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, SK-03601, Martin, Slovak Republic
| | - Maria Brodnanova
- Biomedical Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Michal Pokusa
- Biomedical Center, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Peter Racay
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, SK-03601, Martin, Slovak Republic.
| |
Collapse
|
3
|
Liang W, Huang L, Yuan T, Cheng R, Takahashi Y, Moiseyev GP, Karamichos D, Ma JX. A Method for Real-Time Assessment of Mitochondrial Respiration Using Murine Corneal Biopsy. Invest Ophthalmol Vis Sci 2023; 64:33. [PMID: 37642632 PMCID: PMC10476441 DOI: 10.1167/iovs.64.11.33] [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: 03/17/2023] [Accepted: 07/09/2023] [Indexed: 08/31/2023] Open
Abstract
Purpose To develop and optimize a method to monitor real-time mitochondrial function by measuring the oxygen consumption rate (OCR) in murine corneal biopsy punches with a Seahorse extracellular flux analyzer. Methods Murine corneal biopsies were obtained using a biopsy punch immediately after euthanasia. The corneal metabolic profile was assessed using a Seahorse XFe96 pro analyzer, and mitochondrial respiration was analyzed with specific settings. Results Real-time adenosine triphosphate rate assay showed that mitochondrial oxidative phosphorylation is a major source of adenosine triphosphate production in ex vivo live murine corneal biopsies. Euthanasia methods (carbon dioxide asphyxiation vs. overdosing on anesthetic drugs) did not affect corneal OCR values. Mouse corneal biopsy punches in 1.5-mm diameter generated higher and more reproducible OCR values than those in 1.0-mm diameter. The biopsy punches from the central and off-central cornea did not show significant differences in OCR values. There was no difference in OCR reading by the tissue orientations (the epithelium side up vs. the endothelium side up). No significant differences were found in corneal OCR levels between sexes, strains (C57BL/6J vs. BALB/cJ), or ages (4, 8, and 32 weeks). Using this method, we showed that the wound healing process in the mouse cornea affected mitochondrial activity. Conclusions The present study validated a new strategy to measure real-time mitochondrial function in fresh mouse corneal tissues. This procedure should be helpful for studies of the ex vivo live corneal metabolism in response to genetic manipulations, disease conditions, or pharmacological treatments in mouse models.
Collapse
Affiliation(s)
- Wentao Liang
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Li Huang
- Department of Ophthalmology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Tian Yuan
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Rui Cheng
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Yusuke Takahashi
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Gennadiy P. Moiseyev
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| |
Collapse
|
4
|
Almikhlafi MA, Karami MM, Jana A, Alqurashi TM, Majrashi M, Alghamdi BS, Ashraf GM. Mitochondrial Medicine: A Promising Therapeutic Option Against Various Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:1165-1183. [PMID: 36043795 PMCID: PMC10286591 DOI: 10.2174/1570159x20666220830112408] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/05/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022] Open
Abstract
Abnormal mitochondrial morphology and metabolic dysfunction have been observed in many neurodegenerative disorders (NDDs). Mitochondrial dysfunction can be caused by aberrant mitochondrial DNA, mutant nuclear proteins that interact with mitochondria directly or indirectly, or for unknown reasons. Since mitochondria play a significant role in neurodegeneration, mitochondriatargeted therapies represent a prosperous direction for the development of novel drug compounds that can be used to treat NDDs. This review gives a brief description of how mitochondrial abnormalities lead to various NDDs such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. We further explore the promising therapeutic effectiveness of mitochondria- directed antioxidants, MitoQ, MitoVitE, MitoPBN, and dimebon. We have also discussed the possibility of mitochondrial gene therapy as a therapeutic option for these NDDs.
Collapse
Affiliation(s)
- Mohannad A. Almikhlafi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Mohammed M. Karami
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ankit Jana
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Thamer M. Alqurashi
- Department of Pharmacology, Faculty of Medicine, Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Majrashi
- Department of Pharmacology, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Badrah S. Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md. Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| |
Collapse
|
5
|
Li M, Tang H, Li Z, Tang W. Emerging Treatment Strategies for Cerebral Ischemia-Reperfusion Injury. Neuroscience 2022; 507:112-124. [PMID: 36341725 DOI: 10.1016/j.neuroscience.2022.10.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Cerebral ischemia-reperfusion injury (CI/RI) injury is a common feature of ischemic stroke which occurs when the blood supply is restored after a period of ischemia in the brain. Reduced blood-flow to the brain during CI/RI compromises neuronal cell health as a result of mitochondrial dysfunction, oxidative stress, cytokine production, inflammation and tissue damage. Reperfusion therapy during CI/RI can restore the blood flow to ischemic regions of brain which are not yet infarcted. The long-term goal of CI/RI therapy is to reduce stroke-related neuronal cell death, disability and mortality. A range of drug and interventional therapies have emerged that can alleviate CI/RI mediated oxidative stress, inflammation and apoptosis in the brain. Herein, we review recent studies on CI/RI interventions for which a mechanism of action has been described and the potential of these therapeutic modalities for future use in the clinic.
Collapse
Affiliation(s)
- Mengxing Li
- College of Acupuncture and Massage (Rehabilitation Medical College), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Heyong Tang
- College of Integrated Chinese and Western Medicine (School of Life Sciences), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zhen Li
- College of Acupuncture and Massage (Rehabilitation Medical College), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wei Tang
- College of Acupuncture and Massage (Rehabilitation Medical College), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
6
|
Lee RHC, Wu CYC, Citadin CT, Couto E Silva A, Possoit HE, Clemons GA, Acosta CH, de la Llama VA, Neumann JT, Lin HW. Activation of Neuropeptide Y2 Receptor Can Inhibit Global Cerebral Ischemia-Induced Brain Injury. Neuromolecular Med 2022; 24:97-112. [PMID: 34019239 PMCID: PMC8606017 DOI: 10.1007/s12017-021-08665-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022]
Abstract
Cardiopulmonary arrest (CA) can greatly impact a patient's life, causing long-term disability and death. Although multi-faceted treatment strategies against CA have improved survival rates, the prognosis of CA remains poor. We previously reported asphyxial cardiac arrest (ACA) can cause excessive activation of the sympathetic nervous system (SNS) in the brain, which contributes to cerebral blood flow (CBF) derangements such as hypoperfusion and, consequently, neurological deficits. Here, we report excessive activation of the SNS can cause enhanced neuropeptide Y levels. In fact, mRNA and protein levels of neuropeptide Y (NPY, a 36-amino acid neuropeptide) in the hippocampus were elevated after ACA-induced SNS activation, resulting in a reduced blood supply to the brain. Post-treatment with peptide YY3-36 (PYY3-36), a pre-synaptic NPY2 receptor agonist, after ACA inhibited NPY release and restored brain circulation. Moreover, PYY3-36 decreased neuroinflammatory cytokines, alleviated mitochondrial dysfunction, and improved neuronal survival and neurological outcomes. Overall, NPY is detrimental during/after ACA, but attenuation of NPY release via PYY3-36 affords neuroprotection. The consequences of PYY3-36 inhibit ACA-induced 1) hypoperfusion, 2) neuroinflammation, 3) mitochondrial dysfunction, 4) neuronal cell death, and 5) neurological deficits. The present study provides novel insights to further our understanding of NPY's role in ischemic brain injury.
Collapse
Affiliation(s)
- Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Cristiane T Citadin
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alexandre Couto E Silva
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Harlee E Possoit
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Garrett A Clemons
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Christina H Acosta
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Victoria A de la Llama
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Jake T Neumann
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA.
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA.
| |
Collapse
|
7
|
Increased Post-Hypoxic Oxidative Stress and Activation of the PERK Branch of the UPR in Trap1-Deficient Drosophila melanogaster Is Abrogated by Metformin. Int J Mol Sci 2021; 22:ijms222111586. [PMID: 34769067 PMCID: PMC8583878 DOI: 10.3390/ijms222111586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022] Open
Abstract
Hypoxia is known to impair mitochondrial and endoplasmic reticulum (ER) homeostasis. Post-hypoxic perturbations of the ER proteostasis result in the accumulation of misfolded/unfolded proteins leading to the activation of the Unfolded Protein Response (UPR). Mitochondrial chaperone TNF receptor-associated protein 1 (TRAP1) is reported to preserve mitochondrial membrane potential and to impede reactive oxygen species (ROS) production thereby protecting cells from ER stress as well as oxidative stress. The first-line antidiabetic drug Metformin has been attributed a neuroprotective role after hypoxia. Interestingly, Metformin has been reported to rescue mitochondrial deficits in fibroblasts derived from a patient carrying a homozygous TRAP1 loss-of-function mutation. We sought to investigate a putative link between Metformin, TRAP1, and the UPR after hypoxia. We assessed post-hypoxic/reperfusion longevity, mortality, negative geotaxis, ROS production, metabolic activity, gene expression of antioxidant proteins, and activation of the UPR in Trap1-deficient flies. Following hypoxia, Trap1 deficiency caused higher mortality and greater impairments in negative geotaxis compared to controls. Similarly, post-hypoxic production of ROS and UPR activation was significantly higher in Trap1-deficient compared to control flies. Metformin counteracted the deleterious effects of hypoxia in Trap1-deficient flies but had no protective effect in wild-type flies. We provide evidence that TRAP1 is crucially involved in the post-hypoxic regulation of mitochondrial/ER stress and the activation of the UPR. Metformin appears to rescue Trap1-deficiency after hypoxia mitigating ROS production and downregulating the pro-apoptotic PERK (protein kinase R-like ER kinase) arm of the UPR.
Collapse
|
8
|
Nirody JA, Budin I, Rangamani P. ATP synthase: Evolution, energetics, and membrane interactions. J Gen Physiol 2021; 152:152111. [PMID: 32966553 PMCID: PMC7594442 DOI: 10.1085/jgp.201912475] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/24/2020] [Indexed: 12/24/2022] Open
Abstract
The synthesis of ATP, life’s “universal energy currency,” is the most prevalent chemical reaction in biological systems and is responsible for fueling nearly all cellular processes, from nerve impulse propagation to DNA synthesis. ATP synthases, the family of enzymes that carry out this endless task, are nearly as ubiquitous as the energy-laden molecule they are responsible for making. The F-type ATP synthase (F-ATPase) is found in every domain of life and has facilitated the survival of organisms in a wide range of habitats, ranging from the deep-sea thermal vents to the human intestine. Accordingly, there has been a large amount of work dedicated toward understanding the structural and functional details of ATP synthases in a wide range of species. Less attention, however, has been paid toward integrating these advances in ATP synthase molecular biology within the context of its evolutionary history. In this review, we present an overview of several structural and functional features of the F-type ATPases that vary across taxa and are purported to be adaptive or otherwise evolutionarily significant: ion channel selectivity, rotor ring size and stoichiometry, ATPase dimeric structure and localization in the mitochondrial inner membrane, and interactions with membrane lipids. We emphasize the importance of studying these features within the context of the enzyme’s particular lipid environment. Just as the interactions between an organism and its physical environment shape its evolutionary trajectory, ATPases are impacted by the membranes within which they reside. We argue that a comprehensive understanding of the structure, function, and evolution of membrane proteins—including ATP synthase—requires such an integrative approach.
Collapse
Affiliation(s)
- Jasmine A Nirody
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY.,All Souls College, University of Oxford, Oxford, UK
| | - Itay Budin
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA
| |
Collapse
|
9
|
Al Rahim M, Thatipamula S, Pasinetti GM, Hossain MA. Neuronal Pentraxin 1 Promotes Hypoxic-Ischemic Neuronal Injury by Impairing Mitochondrial Biogenesis via Interactions With Active Bax[6A7] and Mitochondrial Hexokinase II. ASN Neuro 2021; 13:17590914211012888. [PMID: 34098747 PMCID: PMC8191073 DOI: 10.1177/17590914211012888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Mitochondrial dysfunction is a key mechanism of cell death in hypoxic-ischemic brain injury. Neuronal pentraxin 1 (NP1) has been shown to play crucial roles in mitochondria-mediated neuronal death. However, the underlying mechanism(s) of NP1-induced mitochondrial dysfunction in hypoxia-ischemia (HI) remains obscure. Here, we report that NP1 induction following HI and its subsequent localization to mitochondria, leads to disruption of key regulatory proteins for mitochondrial biogenesis. Brain mitochondrial DNA (mtDNA) content and mtDNA-encoded subunit I of complex IV (mtCOX-1) expression was increased post-HI, but not the nuclear DNA-encoded subunit of complex II (nSDH-A). Up-regulation of mitochondrial proteins COXIV and HSP60 further supported enhanced mtDNA function. NP1 interaction with active Bax (Bax6A7) was increased in the brain after HI and in oxygen-glucose deprivation (OGD)-induced neuronal cultures. Importantly, NP1 colocalized with mitochondrial hexokinase II (mtHKII) following OGD leading to HKII dissociation from mitochondria. Knockdown of NP1 or SB216763, a GSK-3 inhibitor, prevented OGD-induced mtHKII dissociation and cellular ATP decrease. NP1 also modulated the expression of mitochondrial transcription factor A (Tfam) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), regulators of mitochondrial biogenesis, following HI. Together, we reveal crucial roles of NP1 in mitochondrial biogenesis involving interactions with Bax[6A7] and mtHKII in HI brain injury.
Collapse
Affiliation(s)
- Md Al Rahim
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Shabarish Thatipamula
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Giulio M Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, United States.,James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States
| | - Mir Ahamed Hossain
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
10
|
Cherevatenko RF, Antsiferov OV, Skachilova SY, Pokrovsky MV, Gureev VV, Banchuk II, Banchuk AY, Golubinskaya MI, Syromyatnikova AA, Rozhkov IS, Mostovykh AA. THE SEARCH FOR NEUROPROTECTIVE COMPOUNDS AMONG NEW ETHYLTHIADIAZOLE DERIVATIVES. PHARMACY & PHARMACOLOGY 2021. [DOI: 10.19163/2307-9266-2019-8-4-263-272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The aim of the study is to search compounds with neuroprotective properties among new ethylthiadiazole derivatives in simulated traumatic brain injury.Materials and methods. The experiment was carried out on 78 white male rats 270±20 g line “Wistar” 5–6 months of age and 120 outbred sexually mature mice weighing 20±2 grams. The article describes the search for compounds with neuroprotective properties among new ethylthiadiazole derivatives under the codes LKHT 4–15, LKHT 10–18, LKHT 11–18, and LKHT 12–18 in experimental traumatic brain injury in rats. Acute toxicity of the compounds was studied. Pharmacological screening was performed using behavioral and neurological research methods. The McGraw stroke score scale modified by I.V. Gannushkina and the mNSS psychometric scale were used in the study. The open field and Rota-rod tests were used to assess the behavioral status of the animals.Results. The compound-LKHT 12–18 at a dose of 50 mg/kg was detected as a leader. In pharmacological correction of pathology, this compound had the lowest percentage of fatality among the studied compounds (8%), the severity of neurological deficit was significantly reduced, the lowest scores and a higher level of motor activity of the limbs were registered. The number of rearing in the group of animals receiving the compound LKHT 12–18 at the dose of 50 mg/kg increased by 1.5 times, statistically significant (p<0.05) in comparison with the control group. Based on the results of the “Rota-rod” test, the total time of holding animals on the rod for 3 attempts was statistically significantly different in the groups administered with LKHT 12–18 derivatives (1.5 times longer) at the dose of 50 mg/kg compared with the control (p<0.05).Conclusion. Based on the results obtained in this study, it is planned to study in more detail the compound LKHT 12–18 at the dose of 50 mg/kg.
Collapse
Affiliation(s)
| | | | - S. Y. Skachilova
- Russian Scientific Center for the Safety of Biologically Active Substances
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Son SH, In YN, Md, Park JS, You Y, Min JH, Yoo I, Cho YC, Jeong W, Ahn HJ, Kang C, Lee BK. Cerebrospinal Fluid Lactate Levels, Brain Lactate Metabolism and Neurologic Outcome in Patients with Out-of-Hospital Cardiac Arrest. Neurocrit Care 2021; 35:262-270. [PMID: 33432527 DOI: 10.1007/s12028-020-01181-1] [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: 08/04/2020] [Accepted: 12/15/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND/OBJECTIVE Cerebrospinal fluid (CSF) and serum lactate levels were assessed to predict poor neurologic outcome 3 months after return of spontaneous circulation (ROSC). We compared arterio-CSF differences in the lactate (ACDL) levels between two neurologic outcome groups. METHODS This retrospective observational study involved out-of-hospital cardiac arrest (OHCA) survivors who had undergone target temperature management. CSF and serum samples were obtained immediately (lactate0), and at 24 (lactate24), 48 (lactate48), and 72 (lactate72) h after ROSC, and ACDL was calculated at each time point. The primary outcome was poor 3-month neurologic outcome (cerebral performance categories 3-5). RESULTS Of 45 patients, 27 (60.0%) showed poor neurologic outcome. At each time point, CSF lactate levels were significantly higher in the poor neurologic outcome group than in the good neurologic outcome group (6.97 vs. 3.37, 4.20 vs. 2.10, 3.50 vs. 2.00, and 2.79 vs. 2.06, respectively; all P < 0.05). CSF lactate's prognostic performance was higher than serum lactate at each time point, and lactate24 showed the highest AUC values (0.89, 95% confidence interval, 0.75-0.97). Over time, ACDL decreased from - 1.30 (- 2.70-0.77) to - 1.70 (- 3.2 to - 0.57) in the poor neurologic outcome group and increased from - 1.22 (- 2.42-0.32) to - 0.64 (- 2.31-0.15) in the good neurologic outcome group. CONCLUSIONS At each time point, CSF lactate showed better prognostic performance than serum lactate. CSF lactate24 showed the highest prognostic performance for 3-month poor neurologic outcome. Over time, ACDL decreased in the poor neurologic outcome group and increased in the good neurologic outcome group.
Collapse
Affiliation(s)
- Seung Ha Son
- Department of Emergency Medicine, Chungnam National University Hospital, 282, Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
| | | | - Md
- Department of Emergency Medicine, Chungnam National University Sejong Hospital, 20, Bodeum 7-ro, Sejong, Republic of Korea
| | - Jung Soo Park
- Department of Emergency Medicine, Chungnam National University Hospital, 282, Munhwa-ro, Jung-gu, Daejeon, Republic of Korea. .,Department of Emergency Medicine, College of Medicine, Chungnam National University, 282, Mokdong-ro, Jung-gu, Daejeon, Republic of Korea.
| | - Yeonho You
- Department of Emergency Medicine, Chungnam National University Hospital, 282, Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
| | - Jin Hong Min
- Department of Emergency Medicine, Chungnam National University Sejong Hospital, 20, Bodeum 7-ro, Sejong, Republic of Korea.,Department of Emergency Medicine, College of Medicine, Chungnam National University, 282, Mokdong-ro, Jung-gu, Daejeon, Republic of Korea
| | - Insool Yoo
- Department of Emergency Medicine, Chungnam National University Hospital, 282, Munhwa-ro, Jung-gu, Daejeon, Republic of Korea.,Department of Emergency Medicine, College of Medicine, Chungnam National University, 282, Mokdong-ro, Jung-gu, Daejeon, Republic of Korea
| | - Yong Chul Cho
- Department of Emergency Medicine, Chungnam National University Hospital, 282, Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
| | - Wonjoon Jeong
- Department of Emergency Medicine, Chungnam National University Hospital, 282, Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
| | - Hong Joon Ahn
- Department of Emergency Medicine, Chungnam National University Hospital, 282, Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
| | - Changshin Kang
- Department of Emergency Medicine, Chungnam National University Hospital, 282, Munhwa-ro, Jung-gu, Daejeon, Republic of Korea
| | - Byung Kook Lee
- Department of Emergency Medicine, Chonnam National University Medical School, Gwangju, 61469, Korea
| |
Collapse
|
12
|
Khatri DK, Choudhary M, Sood A, Singh SB. Anxiety: An ignored aspect of Parkinson’s disease lacking attention. Biomed Pharmacother 2020; 131:110776. [DOI: 10.1016/j.biopha.2020.110776] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/08/2020] [Accepted: 09/17/2020] [Indexed: 12/25/2022] Open
|
13
|
Biswas S, Das H, Das U, Sengupta A, Dey Sharma R, Biswas SC, Dey S. Smokeless tobacco induces toxicity and apoptosis in neuronal cells: a mechanistic evaluation. Free Radic Res 2020; 54:477-496. [PMID: 32842814 DOI: 10.1080/10715762.2020.1805446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Smokeless tobacco (SLT) or chewing tobacco has been a highly addictive practice in India across ages, posing major threat to the systemic health and possibly neurodegeneration. Earlier studies showed components of SLT could be harmful to neuronal health. However, mechanism of SLT in neurodegeneration remained unexplored. This study investigated the detrimental role of SLT on differentiated neuronal cell lines, PC12 and SH-SY5Y by using graded doses of water soluble lyophilised SLT. Reduced cell viability, compromised mitochondrial structure and functions were observed when neuronal cell lines were treated with SLT (6 mg/mL) for 24 h. There was reduction of oxidative phosphorylation and aerobic glycolysis as determined by diminution of ATP production (2.5X) and basal respiration (1.9X). Mitochondrial membrane potential was dropped by 3.5 times. Bid, a pro-apoptotic Bcl-2 family protein, has imperative role in regulating mitochondrial outer membrane permeabilization and subsequent cytochrome c release leading to apoptosis. This article for the first time indicated the involvement of Bid in SLT mediated neurotoxicity and possibly neurodegeneration. SLT treatment enhanced expression of cleaved-Bid in time dependent manner. The involvement of Bid was further confirmed by using Bid specific shRNA which reversed the effects of SLT and conferred significant protection from apoptosis up to 72 h. Thus, our results clearly indicated that SLT induced neuronal cell death occurred via production of ROS, alteration of mitochondrial morphology, membrane potential and oxidative phosphorylation, inactivation of survival pathway and activation of apoptotic markers mediated by Bid. Therefore, Bid could be a potential future therapeutic target for SLT induced neurodegeneration.
Collapse
Affiliation(s)
- Sushobhan Biswas
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Hrishita Das
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Ujjal Das
- Department of Physiology, University of Calcutta, Kolkata, India
| | - Aaveri Sengupta
- Department of Physiology, University of Calcutta, Kolkata, India
| | | | - Subhas Chandra Biswas
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sanjit Dey
- Department of Physiology, University of Calcutta, Kolkata, India
| |
Collapse
|
14
|
Hill RL, Singh IN, Wang JA, Kulbe JR, Hall ED. Protective effects of phenelzine administration on synaptic and non-synaptic cortical mitochondrial function and lipid peroxidation-mediated oxidative damage following TBI in young adult male rats. Exp Neurol 2020; 330:113322. [PMID: 32325157 DOI: 10.1016/j.expneurol.2020.113322] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) results in mitochondrial dysfunction and induction of lipid peroxidation (LP). Lipid peroxidation-derived neurotoxic aldehydes such as 4-HNE and acrolein bind to mitochondrial proteins, inducing additional oxidative damage and further exacerbating mitochondrial dysfunction and LP. Mitochondria are heterogeneous, consisting of both synaptic and non-synaptic populations, with synaptic mitochondria being more vulnerable to injury-dependent consequences. The goal of these studies was to explore the hypothesis that interrupting secondary oxidative damage following TBI using phenelzine (PZ), an aldehyde scavenger, would preferentially protect synaptic mitochondria against LP-mediated damage in a dose- and time-dependent manner. Male Sprague-Dawley rats received a severe (2.2 mm) controlled cortical impact (CCI)-TBI. PZ (3-30 mg/kg) was administered subcutaneously (subQ) at different times post-injury. We found PZ treatment preserves both synaptic and non-synaptic mitochondrial bioenergetics at 24 h and that this protection is partially maintained out to 72 h post-injury using various dosing regimens. The results from these studies indicate that the therapeutic window for the first dose of PZ is likely within the first hour after injury, and the window for administration of the second dose seems to fall between 12 and 24 h. Administration of PZ was able to significantly improve mitochondrial respiration compared to vehicle-treated animals across various states of respiration for both the non-synaptic and synaptic mitochondria. The synaptic mitochondria appear to respond more robustly to PZ treatment than the non-synaptic, and further experimentation will need to be done to further understand these effects in the context of TBI.
Collapse
Affiliation(s)
- Rachel L Hill
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America.
| | - Indrapal N Singh
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America; Department of Neuroscience, 741 S. Limestone St, Lexington, KY 40536-0509, United States of America
| | - Juan A Wang
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America
| | - Jacqueline R Kulbe
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America
| | - Edward D Hall
- University of Kentucky, Spinal Cord and Brain Injury Research Center (SCoBIRC), United States of America; Department of Neuroscience, 741 S. Limestone St, Lexington, KY 40536-0509, United States of America
| |
Collapse
|
15
|
Resuscitation with Drag Reducing Polymers after Traumatic Brain Injury with Hemorrhagic Shock Reduces Microthrombosis and Oxidative Stress. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 31893392 DOI: 10.1007/978-3-030-34461-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Outcome after traumatic brain injury (TBI) is worsened by hemorrhagic shock (HS); however, the existing volume expansion approach with resuscitation fluids (RF) is controversial as it does not adequately alleviate impaired microvascular cerebral blood flow (mCBF). We previously reported that resuscitation fluid with drag reducing polymers (DRP-RF) improves CBF by rheological modulation of hemodynamics. Here, we evaluate the efficacy of DRP-RF, compared to lactated Ringers resuscitation fluid (LR-RF), in reducing cerebral microthrombosis and reperfusion mitochondrial oxidative stress after TBI complicated by HS. Fluid percussion TBI (1.5 ATA, 50 ms) was induced in rats and followed by controlled HS to a mean arterial pressure (MAP) of 40 mmHg. DRP-RF or LR-RF was infused to restore MAP to 60 mmHg for 1 h (pre-hospital period), followed by blood re-infusion to a MAP = 70 mmHg (hospital period). In vivo 2-photon laser scanning microscopy over the parietal cortex was used to monitor microvascular blood flow, nicotinamide adenine dinucleotide (NADH) for tissue oxygen supply and mitochondrial oxidative stress (superoxide by i.v. hydroethidine [HEt], 1 mg/kg) for 4 h after TBI/HS, followed by Dil vascular painting during perfusion-fixation. TBI/HS decreased mCBF resulting in capillary microthrombosis and tissue hypoxia. Microvascular CBF and tissue oxygenation were significantly improved in the DRP-RF compared to the LR-RF treated group (p < 0.05). Reperfusion-induced oxidative stress, reflected by HEt fluorescence, was 32 ± 6% higher in LR-RF vs. DRP-RF (p < 0.05). Post-mortem whole-brain visualization of DiI painted vessels revealed multiple microthromboses in both hemispheres that were 29 ± 3% less in DRP-RF vs. LR-RF group (p < 0.05). Resuscitation after TBI/HS using DRP-RF effectively restores mCBF, reduces hypoxia, microthrombosis formation, and mitochondrial oxidative stress compared to conventional volume expansion with LR-RF.
Collapse
|
16
|
Omelchenko A, Shrirao AB, Bhattiprolu AK, Zahn JD, Schloss RS, Dickson S, Meaney DF, Boustany NN, Yarmush ML, Firestein BL. Dynamin and reverse-mode sodium calcium exchanger blockade confers neuroprotection from diffuse axonal injury. Cell Death Dis 2019; 10:727. [PMID: 31562294 PMCID: PMC6765020 DOI: 10.1038/s41419-019-1908-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 08/20/2019] [Accepted: 08/27/2019] [Indexed: 12/18/2022]
Abstract
Mild traumatic brain injury (mTBI) is a frequently overlooked public health concern that is difficult to diagnose and treat. Diffuse axonal injury (DAI) is a common mTBI neuropathology in which axonal shearing and stretching induces breakdown of the cytoskeleton, impaired axonal trafficking, axonal degeneration, and cognitive dysfunction. DAI is becoming recognized as a principal neuropathology of mTBI with supporting evidence from animal model, human pathology, and neuroimaging studies. As mitochondrial dysfunction and calcium overload are critical steps in secondary brain and axonal injury, we investigated changes in protein expression of potential targets following mTBI using an in vivo controlled cortical impact model. We show upregulated expression of sodium calcium exchanger1 (NCX1) in the hippocampus and cortex at distinct time points post-mTBI. Expression of dynamin-related protein1 (Drp1), a GTPase responsible for regulation of mitochondrial fission, also changes differently post-injury in the hippocampus and cortex. Using an in vitro model of DAI previously reported by our group, we tested whether pharmacological inhibition of NCX1 by SN-6 and of dynamin1, dynamin2, and Drp1 by dynasore mitigates secondary damage. Dynasore and SN-6 attenuate stretch injury-induced swelling of axonal varicosities and mitochondrial fragmentation. In addition, we show that dynasore, but not SN-6, protects against H2O2-induced damage in an organotypic oxidative stress model. As there is currently no standard treatment to mitigate cell damage induced by mTBI and DAI, this work highlights two potential therapeutic targets for treatment of DAI in multiple models of mTBI and DAI.
Collapse
Affiliation(s)
- Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Anil B Shrirao
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Atul K Bhattiprolu
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Jeffrey D Zahn
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Samantha Dickson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104-6391, USA
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104-6391, USA
| | - Nada N Boustany
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854-8082, USA.
| |
Collapse
|
17
|
Duan X, Li L, Gan J, Peng C, Wang X, Chen W, Peng D. Identification and functional analysis of circular RNAs induced in rats by middle cerebral artery occlusion. Gene 2019; 701:139-145. [DOI: 10.1016/j.gene.2019.03.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/05/2019] [Accepted: 03/22/2019] [Indexed: 01/01/2023]
|
18
|
Arambula SE, Reinl EL, El Demerdash N, McCarthy MM, Robertson CL. Sex differences in pediatric traumatic brain injury. Exp Neurol 2019; 317:168-179. [PMID: 30831070 DOI: 10.1016/j.expneurol.2019.02.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
Abstract
The response of the developing brain to traumatic injury is different from the response of the mature, adult brain. There are critical developmental trajectories in the young brain, whereby injury can lead to long term functional abnormalities. Emerging preclinical and clinical literature supports the presence of significant sex differences in both the response to and the recovery from pediatric traumatic brain injury (TBI). These sex differences are seen at all pediatric ages, including neonates/infants, pre-pubertal children, and adolescents. As importantly, the response to neuroprotective therapies or treatments can differ between male and females subjects. These sex differences can result from several biologic origins, and may manifest differently during the various phases of brain and body development. Recognizing and understanding these potential sex differences is crucial, and should be considered in both preclinical and clinical studies of pediatric TBI.
Collapse
Affiliation(s)
- Sheryl E Arambula
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Erin L Reinl
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nagat El Demerdash
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Margaret M McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Courtney L Robertson
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
19
|
Dong Y, Hu XH, Wu T, Wang T. Effect of hyperbaric oxygenation therapy on post-concussion syndrome. Exp Ther Med 2018; 16:2193-2202. [PMID: 30186458 PMCID: PMC6122203 DOI: 10.3892/etm.2018.6463] [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: 12/25/2016] [Accepted: 07/11/2017] [Indexed: 11/12/2022] Open
Abstract
The present review evaluated the effect of hyperbaric oxygenation (HBO) therapy on post-concussion syndrome (PCS). Searches for publications from the earliest date possible up until the first week of 2016 were conducted using the electronic databases Cochrane, EBSCOhost, Embase, Ovid MEDLINE, PubMed and Web of Science. Additional trials were identified through reference list scanning. Randomized controlled trials assessing the effectiveness of HBO therapy in PCS were selected and tested for eligibility for inclusion in the present review. Two independent reviewers conducted data extraction and the Cochrane Collaboration's recommended method was used to assess the risk of bias in each study included. Review Manager 5.3 software was used for data synthesis and analysis and the standardized mean difference (SMD) or mean difference (MD) was estimated with a fixed or random effects model using a 95% confidence interval (CI). A total of 127 articles were identified, 4 of which were eligible for final analysis. The meta-analysis identified no difference in the Rivermead Post-Concussion Symptoms Questionnaire (MD=1.23; 95% CI, -3.47-5.94; P>0.05; I2=35%) or Post-Traumatic Stress Disorder Checklist (PCL) scores (SMD=0.12; 95% CI, -0.31-0.54; P>0.05; I2=0%) scores between groups receiving different oxygen doses. The differences in PCL scores (SMD=-0.13, 95% CI, -0.80-0.53; P>0.05; I2=63%) and neurobehavioral symptoms (SMD=-1.00, 95% CI, -2.58-0.58; P>0.05; I2=92%) between the HBO and sham groups were not significant. The current study demonstrated that HBO therapy has no significant effect on PCS compared with the sham group. Therefore, it was determined that effective design and execution of a large clinical trial, which includes treatment, control and sham groups is required in the future.
Collapse
Affiliation(s)
- Yang Dong
- Department of Rehabilitation Medicine, Hangzhou Hospital of Zhejiang CAPF, Hangzhou, Zhejiang 310016, P.R. China
- Institute of Rehabilitation Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xia Hua Hu
- Department of Rehabilitation Medicine, Hangzhou Hospital of Zhejiang CAPF, Hangzhou, Zhejiang 310016, P.R. China
| | - Tao Wu
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Tong Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
20
|
Jaber SM, Bordt EA, Bhatt NM, Lewis DM, Gerecht S, Fiskum G, Polster BM. Sex differences in the mitochondrial bioenergetics of astrocytes but not microglia at a physiologically relevant brain oxygen tension. Neurochem Int 2018; 117:82-90. [PMID: 28888963 PMCID: PMC5839942 DOI: 10.1016/j.neuint.2017.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 08/14/2017] [Accepted: 09/05/2017] [Indexed: 11/22/2022]
Abstract
Biological sex is thought to influence mitochondrial bioenergetic function. Previous respiration measurements examining brain mitochondrial sex differences were made at atmospheric oxygen using isolated brain mitochondria. Oxygen is 160 mm Hg (21%) in the atmosphere, while the oxygen tension in the brain generally ranges from ∼5 to 45 mm Hg (∼1-6% O2). This study tested the hypothesis that sex and/or brain physiological oxygen tension influence the mitochondrial bioenergetic properties of primary rat cortical astrocytes and microglia. Oxygen consumption was measured with a Seahorse XF24 cell respirometer in an oxygen-controlled environmental chamber. Strikingly, male astrocytes had a higher maximal respiration than female astrocytes when cultured and assayed at 3% O2. Three percent O2 yielded a low physiological dissolved O2 level of ∼1.2% (9.1 mm Hg) at the cell monolayer during culture and 1.2-3.0% O2 during assays. No differences in bioenergetic parameters were observed between male and female astrocytes at 21% O2 (dissolved O2 of ∼19.7%, 150 mm Hg during culture) or between either of these cell populations and female astrocytes at 3% O2. In contrast to astrocytes, microglia showed no sex differences in mitochondrial bioenergetic parameters at either oxygen level, regardless of whether they were non-stimulated or activated to a proinflammatory state. There were also no O2- or sex-dependent differences in proinflammatory TNF-α or IL-1β cytokine secretion measured at 18 h activation. Overall, results reveal an intriguing sex variance in astrocytic maximal respiration that requires additional investigation. Findings also demonstrate that sex differences can be masked by conducting experiments at non-physiological O2.
Collapse
Affiliation(s)
- Sausan M Jaber
- Department of Anesthesiology, and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore ST., MSTF 5-34, Baltimore, MD, 21201, USA; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 685 W. Baltimore ST., MSTF 5-34, Baltimore, MD, 21201, USA
| | - Evan A Bordt
- Department of Anesthesiology, and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore ST., MSTF 5-34, Baltimore, MD, 21201, USA
| | - Niraj M Bhatt
- Department of Anesthesiology, and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore ST., MSTF 5-34, Baltimore, MD, 21201, USA
| | - Daniel M Lewis
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, 3400 North Charles Street, Shaffer Hall 200C, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Institute for NanoBioTechnology, Johns Hopkins University, 3400 North Charles Street, Shaffer Hall 200C, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Shaffer Hall 200C, Baltimore, MD 21218, USA
| | - Gary Fiskum
- Department of Anesthesiology, and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore ST., MSTF 5-34, Baltimore, MD, 21201, USA; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 685 W. Baltimore ST., MSTF 5-34, Baltimore, MD, 21201, USA
| | - Brian M Polster
- Department of Anesthesiology, and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore ST., MSTF 5-34, Baltimore, MD, 21201, USA; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 685 W. Baltimore ST., MSTF 5-34, Baltimore, MD, 21201, USA.
| |
Collapse
|
21
|
Hill RL, Kulbe JR, Singh IN, Wang JA, Hall ED. Synaptic Mitochondria are More Susceptible to Traumatic Brain Injury-induced Oxidative Damage and Respiratory Dysfunction than Non-synaptic Mitochondria. Neuroscience 2018; 386:265-283. [PMID: 29960045 DOI: 10.1016/j.neuroscience.2018.06.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 06/12/2018] [Accepted: 06/18/2018] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) results in mitochondrial dysfunction and induction of lipid peroxidation (LP). Lipid peroxidation-derived neurotoxic aldehydes such as 4-HNE and acrolein bind to mitochondrial proteins, inducing additional oxidative damage and further exacerbating mitochondrial dysfunction and LP. Mitochondria are heterogeneous, consisting of both synaptic and non-synaptic populations. Synaptic mitochondria are reported to be more vulnerable to injury; however, this is the first study to characterize the temporal profile of synaptic and non-synaptic mitochondria following TBI, including investigation of respiratory dysfunction and oxidative damage to mitochondrial proteins between 3 and 120 h following injury. These results indicate that synaptic mitochondria are indeed the more vulnerable population, showing both more rapid and severe impairments than non-synaptic mitochondria. By 24 h, synaptic respiration is significantly impaired compared to synaptic sham, whereas non-synaptic respiration does not decline significantly until 48 h. Decreases in respiration are associated with increases in oxidative damage to synaptic and non-synaptic mitochondrial proteins at 48 h and 72 h, respectively. These results indicate that the therapeutic window for mitochondria-targeted pharmacological neuroprotectants to prevent respiratory dysfunction is shorter for the more vulnerable synaptic mitochondria than for the non-synaptic population.
Collapse
Affiliation(s)
- Rachel L Hill
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Jacqueline R Kulbe
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Indrapal N Singh
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Juan A Wang
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Edward D Hall
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States.
| |
Collapse
|
22
|
Kulbe JR, Singh IN, Wang JA, Cebak JE, Hall ED. Continuous Infusion of Phenelzine, Cyclosporine A, or Their Combination: Evaluation of Mitochondrial Bioenergetics, Oxidative Damage, and Cytoskeletal Degradation following Severe Controlled Cortical Impact Traumatic Brain Injury in Rats. J Neurotrauma 2018; 35:1280-1293. [PMID: 29336204 PMCID: PMC5962911 DOI: 10.1089/neu.2017.5353] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
To date, all monotherapy clinical traumatic brain injury (TBI) trials have failed, and there are currently no Food and Drug Administration (FDA)-approved pharmacotherapies for the acute treatment of severe TBI. Due to the complex secondary injury cascade following injury, there is a need to develop multi-mechanistic combinational neuroprotective approaches for the treatment of acute TBI. As central mediators of the TBI secondary injury cascade, both mitochondria and lipid peroxidation-derived aldehydes make promising therapeutic targets. Cyclosporine A (CsA), an FDA-approved immunosuppressant capable of inhibiting the mitochondrial permeability transition pore, and phenelzine (PZ), an FDA-approved monoamine oxidase inhibitor capable of scavenging neurotoxic lipid peroxidation-derived aldehydes, have both been shown to be partially neuroprotective following experimental TBI. Therefore, it follows that the combination of PZ and CsA may enhance neuroprotection over either agent alone through the combining of distinct but complementary mechanisms of action. Additionally, as the first 72 h represents a critical time period following injury, it follows that continuous drug infusion over the first 72 h following injury may also lead to optimal neuroprotective effects. This is the first study to examine the effects of a 72 h subcutaneous continuous infusion of PZ, CsA, and the combination of these two agents on mitochondrial respiration, mitochondrial bound 4-hydroxynonenal (4-HNE), and acrolein, and α-spectrin degradation 72 h following a severe controlled cortical impact injury in rats. Our results indicate that individually, both CsA and PZ are able to attenuate mitochondrial 4-HNE and acrolein, PZ is able to maintain mitochondrial respiratory control ratio and cytoskeletal integrity but together, PZ and CsA are unable to maintain neuroprotective effects.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord and Brain Injury Research Center and Department of Neuroscience, University of Kentucky College of Medicine , Lexington, Kentucky
| | - Indrapal N Singh
- Spinal Cord and Brain Injury Research Center and Department of Neuroscience, University of Kentucky College of Medicine , Lexington, Kentucky
| | - Juan A Wang
- Spinal Cord and Brain Injury Research Center and Department of Neuroscience, University of Kentucky College of Medicine , Lexington, Kentucky
| | - John E Cebak
- Spinal Cord and Brain Injury Research Center and Department of Neuroscience, University of Kentucky College of Medicine , Lexington, Kentucky
| | - Edward D Hall
- Spinal Cord and Brain Injury Research Center and Department of Neuroscience, University of Kentucky College of Medicine , Lexington, Kentucky
| |
Collapse
|
23
|
Li M, Sirko S. Traumatic Brain Injury: At the Crossroads of Neuropathology and Common Metabolic Endocrinopathies. J Clin Med 2018. [PMID: 29538298 PMCID: PMC5867585 DOI: 10.3390/jcm7030059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Building on the seminal work by Geoffrey Harris in the 1970s, the neuroendocrinology field, having undergone spectacular growth, has endeavored to understand the mechanisms of hormonal connectivity between the brain and the rest of the body. Given the fundamental role of the brain in the orchestration of endocrine processes through interactions among neurohormones, it is thus not surprising that the structural and/or functional alterations following traumatic brain injury (TBI) can lead to endocrine changes affecting the whole organism. Taking into account that systemic hormones also act on the brain, modifying its structure and biochemistry, and can acutely and chronically affect several neurophysiological endpoints, the question is to what extent preexisting endocrine dysfunction may set the stage for an adverse outcome after TBI. In this review, we provide an overview of some aspects of three common metabolic endocrinopathies, e.g., diabetes mellitus, obesity, and thyroid dysfunction, and how these could be triggered by TBI. In addition, we discuss how the complex endocrine networks are woven into the responses to sudden changes after TBI, as well as some of the potential mechanisms that, separately or synergistically, can influence outcomes after TBI.
Collapse
Affiliation(s)
- Melanie Li
- Physiological Genomics, Biomedical Center (BMC), Institute of Physiology, Medical Faculty of the Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany.
| | - Swetlana Sirko
- Physiological Genomics, Biomedical Center (BMC), Institute of Physiology, Medical Faculty of the Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany.
- Institute of Stem Cell Research, Helmholtz Center Munich, Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany.
| |
Collapse
|
24
|
Zhao H, Du H, Liu M, Gao S, Li N, Chao Y, Li R, Chen W, Lou Z, Dong X. Integrative Proteomics-Metabolomics Strategy for Pathological Mechanism of Vascular Depression Mouse Model. J Proteome Res 2017; 17:656-669. [PMID: 29190102 DOI: 10.1021/acs.jproteome.7b00724] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vascular depression (VD), a subtype of depression, is caused by vascular diseases or cerebrovascular risk factors. Recently, the proportion of VD patients has increased significantly, which severely affects their quality of life. However, the current pathogenesis of VD has not yet been fully understood, and the basic research is not adequate. In this study, on the basis of the combination of LC-MS-based proteomics and metabolomics, we aimed to establish a protein metabolism regulatory network in a murine VD model to elucidate a more comprehensive impact of VD on organisms. We detected 44 metabolites and 304 proteins with different levels in the hippocampus samples from VD mice using a combination of metabolomic and proteomics analyses with an isobaric tags for relative and absolute quantification (iTRAQ) method. We constructed a protein-to-metabolic regulatory network by correlating and integrating the differential metabolites and proteins using ingenuity pathway analysis. Then we quantitatively validated the levels of the bimolecules shown in the bioinformatics analysis using LC-MS/MS and Western blotting. Validation results suggested changes in the regulation of neuroplasticity, transport of neurotransmitters, neuronal cell proliferation and apoptosis, and disorders of amino acids, lipids and energy metabolism. These proteins and metabolites involved in these dis-regulated pathways will provide a more targeted and credible direction to study the mechanism of VD. Therefore, this paper presents an approach and strategy that was applied in integrative proteomics and metabolomics for research and screening potential targets and biomarkers of VD, which could be more precise and credible in a field lacking adequate basic research.
Collapse
Affiliation(s)
- Hongxia Zhao
- School of Pharmacy, Second Military Medical University , Shanghai 200433, China
| | - Hongli Du
- Department of Pharmacy, Eastern Hepatobiliary Surgery Hospital , Shanghai 200433, China
| | - Min Liu
- Pharmacy Department of Changhai Hospital, Second Military Medical University , Shanghai 200433, China
| | - Songyan Gao
- School of Pharmacy, Second Military Medical University , Shanghai 200433, China
| | - Na Li
- School of Pharmacy, Second Military Medical University , Shanghai 200433, China
| | - Yufan Chao
- School of Pharmacy, Second Military Medical University , Shanghai 200433, China
| | - Ruiqing Li
- School of Life Sciences and Technology, Shanghai Tech University , Shanghai 200433, China
| | - Wei Chen
- Changhai Hospital, Second Military Medical University , Shanghai 200433, China
| | - Ziyang Lou
- School of Pharmacy, Second Military Medical University , Shanghai 200433, China
| | - Xin Dong
- School of Pharmacy, Second Military Medical University , Shanghai 200433, China
| |
Collapse
|
25
|
Hill RL, Singh IN, Wang JA, Hall ED. Time courses of post-injury mitochondrial oxidative damage and respiratory dysfunction and neuronal cytoskeletal degradation in a rat model of focal traumatic brain injury. Neurochem Int 2017; 111:45-56. [PMID: 28342966 PMCID: PMC5610595 DOI: 10.1016/j.neuint.2017.03.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 03/14/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) results in rapid reactive oxygen species (ROS) production and oxidative damage to essential brain cellular components leading to neuronal dysfunction and cell death. It is increasingly appreciated that a major player in TBI-induced oxidative damage is the reactive nitrogen species (RNS) peroxynitrite (PN) which is produced in large part in injured brain mitochondria. Once formed, PN decomposes into highly reactive free radicals that trigger membrane lipid peroxidation (LP) of polyunsaturated fatty acids (e.g. arachidonic acid) and protein nitration (3-nitrotyrosine, 3-NT) in mitochondria and other cellular membranes causing various functional impairments to mitochondrial oxidative phosphorylation and calcium (Ca2+) buffering capacity. The LP also results in the formation of neurotoxic reactive aldehyde byproducts including 4-hydroxynonenal (4-HNE) and propenal (acrolein) which exacerbates ROS/RNS production and oxidative protein damage in the injured brain. Ultimately, this results in intracellular Ca2+ overload that activates proteolytic degradation of α-spectrin, a neuronal cytoskeletal protein. Therefore, the aim of this study was to establish the temporal evolution of mitochondrial dysfunction, oxidative damage and cytoskeletal degradation in the brain following a severe controlled cortical impact (CCI) TBI in young male adult rats. In mitochondria isolated from an 8 mm diameter cortical punch including the 5 mm wide impact site and their respiratory function studied ex vivo, we observed an initial decrease in complex I and II mitochondrial bioenergetics within 3 h (h). For complex I bioenergetics, this partially recovered by 12-16 h, whereas for complex II respiration the recovery was complete by 12 h. During the first 24 h, there was no evidence of an injury-induced increase in LP or protein nitration in mitochondrial or cellular homogenates. However, beginning at 24 h, there was a gradual secondary decline in complex I and II respiration that peaked at 72 h. post-TBI that coincided with progressive peroxidation of mitochondrial and cellular lipids, protein nitration and protein modification by 4-HNE and acrolein. The oxidative damage and respiratory failure paralleled an increase in Ca2+-induced proteolytic degradation of the neuronal cytoskeletal protein α-spectrin indicating a failure of intracellular Ca2+ homeostasis. These findings of a surprisingly delayed peak in secondary injury, suggest that the therapeutic window and needed treatment duration for certain antioxidant treatment strategies following CCI-TBI in rodents may be longer than previously believed.
Collapse
Affiliation(s)
- Rachel L Hill
- University of Kentucky College of Medicine, Spinal Cord and Brain Injury Research Center (SCoBIRC), 741 S. Limestone St, Lexington, KY 40536-0509, USA
| | - Indrapal N Singh
- University of Kentucky College of Medicine, Spinal Cord and Brain Injury Research Center (SCoBIRC), 741 S. Limestone St, Lexington, KY 40536-0509, USA; University of Kentucky College of Medicine, Department of Neuroscience, 741 S. Limestone St, Lexington, KY 40536-0509, USA
| | - Juan A Wang
- University of Kentucky College of Medicine, Spinal Cord and Brain Injury Research Center (SCoBIRC), 741 S. Limestone St, Lexington, KY 40536-0509, USA
| | - Edward D Hall
- University of Kentucky College of Medicine, Spinal Cord and Brain Injury Research Center (SCoBIRC), 741 S. Limestone St, Lexington, KY 40536-0509, USA; University of Kentucky College of Medicine, Department of Neuroscience, 741 S. Limestone St, Lexington, KY 40536-0509, USA.
| |
Collapse
|
26
|
Kulbe JR, Hall ED. Chronic traumatic encephalopathy-integration of canonical traumatic brain injury secondary injury mechanisms with tau pathology. Prog Neurobiol 2017; 158:15-44. [PMID: 28851546 PMCID: PMC5671903 DOI: 10.1016/j.pneurobio.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
In recent years, a new neurodegenerative tauopathy labeled Chronic Traumatic Encephalopathy (CTE), has been identified that is believed to be primarily a sequela of repeated mild traumatic brain injury (TBI), often referred to as concussion, that occurs in athletes participating in contact sports (e.g. boxing, American football, Australian football, rugby, soccer, ice hockey) or in military combatants, especially after blast-induced injuries. Since the identification of CTE, and its neuropathological finding of deposits of hyperphosphorylated tau protein, mechanistic attention has been on lumping the disorder together with various other non-traumatic neurodegenerative tauopathies. Indeed, brains from suspected CTE cases that have come to autopsy have been confirmed to have deposits of hyperphosphorylated tau in locations that make its anatomical distribution distinct for other tauopathies. The fact that these individuals experienced repetitive TBI episodes during their athletic or military careers suggests that the secondary injury mechanisms that have been extensively characterized in acute TBI preclinical models, and in TBI patients, including glutamate excitotoxicity, intracellular calcium overload, mitochondrial dysfunction, free radical-induced oxidative damage and neuroinflammation, may contribute to the brain damage associated with CTE. Thus, the current review begins with an in depth analysis of what is known about the tau protein and its functions and dysfunctions followed by a discussion of the major TBI secondary injury mechanisms, and how the latter have been shown to contribute to tau pathology. The value of this review is that it might lead to improved neuroprotective strategies for either prophylactically attenuating the development of CTE or slowing its progression.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States
| | - Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States.
| |
Collapse
|
27
|
Shoshan-Barmatz V, Maldonado EN, Krelin Y. VDAC1 at the crossroads of cell metabolism, apoptosis and cell stress. Cell Stress 2017; 1:11-36. [PMID: 30542671 PMCID: PMC6287957 DOI: 10.15698/cst2017.10.104] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review presents current knowledge related to VDAC1 as a multi-functional mitochondrial protein acting on both sides of the coin, regulating cell life and death, and highlighting these functions in relation to disease. It is now recognized that VDAC1 plays a crucial role in regulating the metabolic and energetic functions of mitochondria. The location of VDAC1 at the outer mitochondrial membrane (OMM) allows the control of metabolic cross-talk between mitochondria and the rest of the cell and also enables interaction of VDAC1 with proteins involved in metabolic and survival pathways. Along with regulating cellular energy production and metabolism, VDAC1 is also involved in the process of mitochondria-mediated apoptosis by mediating the release of apoptotic proteins and interacting with anti-apoptotic proteins. VDAC1 functions in the release of apoptotic proteins located in the mitochondrial intermembrane space via oligomerization to form a large channel that allows passage of cytochrome c and AIF and their release to the cytosol, subsequently resulting in apoptotic cell death. VDAC1 also regulates apoptosis via interactions with apoptosis regulatory proteins, such as hexokinase, Bcl2 and Bcl-xL, some of which are also highly expressed in many cancers. This review also provides insight into VDAC1 function in Ca2+ homeostasis, oxidative stress, and presents VDAC1 as a hub protein interacting with over 100 proteins. Such interactions enable VDAC1 to mediate and regulate the integration of mitochondrial functions with cellular activities. VDAC1 can thus be considered as standing at the crossroads between mitochondrial metabolite transport and apoptosis and hence represents an emerging cancer drug target.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Eduardo N Maldonado
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC. USA
| | - Yakov Krelin
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| |
Collapse
|
28
|
Melatonin pre-treatment mitigates SHSY-5Y cells against oxaliplatin induced mitochondrial stress and apoptotic cell death. PLoS One 2017; 12:e0180953. [PMID: 28732061 PMCID: PMC5521772 DOI: 10.1371/journal.pone.0180953] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/23/2017] [Indexed: 01/14/2023] Open
Abstract
Oxaliplatin (Oxa) treatment to SH-SY5Y human neuroblastoma cells has been shown by previous studies to induce oxidative stress, which in turn modulates intracellular signaling cascades resulting in cell death. While this phenomenon of Oxa-induced neurotoxicity is known, the underlying mechanisms involved in this cell death cascade must be clarified. Moreover, there is still little known regarding the roles of neuronal mitochondria and cytosolic compartments in mediating Oxa-induced neurotoxicity. With a better grasp of the mechanisms driving neurotoxicity in Oxa-treated SH-SY5Y cells, we can then identify certain pathways to target in protecting against neurotoxic cell damage. Therefore, the purpose of this study was to determine whether one such agent, melatonin (Mel), could confer protection against Oxa-induced neurotoxicity in SH-SY5Y cells. Results from the present study found Oxa to significantly reduce SH-SY5Y cell viability in a dose-dependent manner. Alternatively, we found Mel pre-treatment to SH-SY5Y cells to attenuate Oxa-induced toxicity, resulting in a markedly increased cell viability. Mel exerted its protective effects by regulating reactive oxygen species (ROS) production and reducing superoxide radicals inside Oxa-exposed. In addition, we observed pre-treatment with Mel to rescue Oxa-treated cells by protecting mitochondria. As Oxa-treatment alone decreases mitochondrial membrane potential (Δψm), resulting in an altered Bcl-2/Bax ratio and release of sequestered cytochrome c, so Mel was shown to inhibit these pathways. Mel was also found to inhibit proteolytic activation of caspase 3, inactivation of Poly (ADP Ribose) polymerase, and DNA damage, thereby allowing SH-SY5Y cells to resist apoptotic cell death. Collectively, our results suggest a role for melatonin in reducing Oxa induced neurotoxicity. Further studies exploring melatonin’s protective effects may prove successful in eliciting pathways to further alter the neurotoxic pathways of platinum compounds in cancer treatment.
Collapse
|
29
|
Bell JE, Seifert JL, Shimizu EN, Sucato DJ, Romero-Ortega MI. Atraumatic Spine Distraction Induces Metabolic Distress in Spinal Motor Neurons. J Neurotrauma 2017; 34:2034-2044. [DOI: 10.1089/neu.2016.4779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Jennifer E.S. Bell
- Bioengineering Department, University of Texas at Dallas, Richardson, Texas
| | | | - Eileen N. Shimizu
- Bioengineering Department, University of Texas at Dallas, Richardson, Texas
| | - Daniel J. Sucato
- Department of Orthopedic Surgery, Texas Scottish Rite Hospital for Children, Dallas, Texas
| | - Mario I. Romero-Ortega
- Bioengineering Department, University of Texas at Dallas, Richardson, Texas
- Surgery Department, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
30
|
Gollihue JL, Patel SP, Mashburn C, Eldahan KC, Sullivan PG, Rabchevsky AG. Optimization of mitochondrial isolation techniques for intraspinal transplantation procedures. J Neurosci Methods 2017; 287:1-12. [PMID: 28554833 DOI: 10.1016/j.jneumeth.2017.05.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Proper mitochondrial function is essential to maintain normal cellular bioenergetics and ionic homeostasis. In instances of severe tissue damage, such as traumatic brain and spinal cord injury, mitochondria become damaged and unregulated leading to cell death. The relatively unexplored field of mitochondrial transplantation following neurotrauma is based on the theory that replacing damaged mitochondria with exogenous respiratory-competent mitochondria can restore overall tissue bioenergetics. NEW METHOD We optimized techniques in vitro to prepare suspensions of isolated mitochondria for transplantation in vivo. Mitochondria isolated from cell culture were genetically labeled with turbo-green fluorescent protein (tGFP) for imaging and tracking purposes in vitro and in vivo. RESULTS We used time-lapse confocal imaging to reveal the incorporation of exogenous fluorescently-tagged mitochondria into PC-12 cells after brief co-incubation. Further, we show that mitochondria can be injected into the spinal cord with immunohistochemical evidence of host cellular uptake within 24h. COMPARISON TO EXISTING METHODS Our methods utilize transgenic fluorescent labeling of mitochondria for a nontoxic and photostable alternative to other labeling methods. Substrate addition to isolated mitochondria helped to restore state III respiration at room temperature prior to transplantation. These experiments delineate refined methods to use transgenic cell lines for the purpose of isolating well coupled mitochondria that have a permanent fluorescent label that allows real time tracking of transplanted mitochondria in vitro, as well as imaging in situ. CONCLUSIONS These techniques lay the foundation for testing the potential therapeutic effects of mitochondrial transplantation following spinal cord injury and other animal models of neurotrauma.
Collapse
Affiliation(s)
- Jenna L Gollihue
- University of Kentucky, Department of Physiology, Lexington, KY 40536-0509, United States; University of Kentucky, Spinal Cord & Brain Injury Research Center, Lexington, KY 40536-0509, United States
| | - Samir P Patel
- University of Kentucky, Department of Physiology, Lexington, KY 40536-0509, United States; University of Kentucky, Spinal Cord & Brain Injury Research Center, Lexington, KY 40536-0509, United States
| | - Charlie Mashburn
- University of Kentucky, Spinal Cord & Brain Injury Research Center, Lexington, KY 40536-0509, United States
| | - Khalid C Eldahan
- University of Kentucky, Department of Physiology, Lexington, KY 40536-0509, United States; University of Kentucky, Spinal Cord & Brain Injury Research Center, Lexington, KY 40536-0509, United States
| | - Patrick G Sullivan
- University of Kentucky, Department of Neuroscience, Lexington, KY 40536-0509, United States; University of Kentucky, Spinal Cord & Brain Injury Research Center, Lexington, KY 40536-0509, United States
| | - Alexander G Rabchevsky
- University of Kentucky, Department of Physiology, Lexington, KY 40536-0509, United States; University of Kentucky, Spinal Cord & Brain Injury Research Center, Lexington, KY 40536-0509, United States.
| |
Collapse
|
31
|
Bakthavachalam P, Shanmugam PST. Mitochondrial dysfunction - Silent killer in cerebral ischemia. J Neurol Sci 2017; 375:417-423. [PMID: 28320180 DOI: 10.1016/j.jns.2017.02.043] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/27/2017] [Accepted: 02/20/2017] [Indexed: 10/20/2022]
Abstract
Mitochondrial dysfunction aggravates ischemic neuronal injury through activation of various pathophysiological and molecular mechanisms. Ischemic neuronal injury is particularly intensified during reperfusion due to impairment of mitochondrial function. Mitochondrial mutilation instigates alterations in calcium homeostasis in neurons, which plays a pivotal role in the maintenance of normal neuronal function. Increase in intracellular calcium level in mitochondria triggers the opening of mitochondrial transition pore and over production of reactive oxygen species (ROS). Several investigations have concluded that ROS not only contribute to lipids and proteins damage, but also transduce apoptotic signals leading to neuronal death. In addition to the above mentioned reasons, endoplasmic reticulum (ER) stress due to excitotoxicity also leads to neuronal death. Recently, some newer proteins have been claimed to induce "mitophagy" by triggering the receptors on autophagic membranes leading to neurodegeneration. This review summarizes the mechanisms underlying neuronal death involving mitochondrial dysfunction and mitophagy.
Collapse
Affiliation(s)
- Pramila Bakthavachalam
- Sri Ramachandra University, No. 1, Ramachandra Nagar, Porur, Chennai, Tamil Nadu, India.
| | | |
Collapse
|
32
|
Mitochondrial Calcium Uptake in Activation of the Permeability Transition Pore and Cell Death. MOLECULAR BASIS FOR MITOCHONDRIAL SIGNALING 2017. [DOI: 10.1007/978-3-319-55539-3_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Ma X, Xie Y, Chen Y, Han B, Li J, Qi S. Post-ischemia mdivi-1 treatment protects against ischemia/reperfusion-induced brain injury in a rat model. Neurosci Lett 2016; 632:23-32. [DOI: 10.1016/j.neulet.2016.08.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/06/2016] [Accepted: 08/15/2016] [Indexed: 01/05/2023]
|
34
|
Sinha S, Raheja A, Samson N, Bhoi S, Selvi A, Sharma P, Sharma BS. Blood mitochondrial enzymatic assay as a predictor of long-term outcome in severe traumatic brain injury. J Clin Neurosci 2016; 30:31-38. [DOI: 10.1016/j.jocn.2015.10.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/23/2015] [Accepted: 10/25/2015] [Indexed: 02/07/2023]
|
35
|
Scaini G, Rezin GT, Carvalho AF, Streck EL, Berk M, Quevedo J. Mitochondrial dysfunction in bipolar disorder: Evidence, pathophysiology and translational implications. Neurosci Biobehav Rev 2016; 68:694-713. [PMID: 27377693 DOI: 10.1016/j.neubiorev.2016.06.040] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 06/26/2016] [Accepted: 06/30/2016] [Indexed: 01/05/2023]
Abstract
Bipolar disorder (BD) is a chronic psychiatric illness characterized by severe and biphasic changes in mood. Several pathophysiological mechanisms have been hypothesized to underpin the neurobiology of BD, including the presence of mitochondrial dysfunction. A confluence of evidence points to an underlying dysfunction of mitochondria, including decreases in mitochondrial respiration, high-energy phosphates and pH; changes in mitochondrial morphology; increases in mitochondrial DNA polymorphisms; and downregulation of nuclear mRNA molecules and proteins involved in mitochondrial respiration. Mitochondria play a pivotal role in neuronal cell survival or death as regulators of both energy metabolism and cell survival and death pathways. Thus, in this review, we discuss the genetic and physiological components of mitochondria and the evidence for mitochondrial abnormalities in BD. The final part of this review discusses mitochondria as a potential target of therapeutic interventions in BD.
Collapse
Affiliation(s)
- Giselli Scaini
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Laboratory of Bioenergetics, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gislaine T Rezin
- Laboratory of Clinical and Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Sul de Santa Catarina, Tubarão, SC, Brazil
| | - Andre F Carvalho
- Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Emilio L Streck
- Laboratory of Bioenergetics, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, Faculty of Health, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health and The Centre for Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
36
|
Kiss DS, Toth I, Jocsak G, Sterczer A, Bartha T, Frenyo LV, Zsarnovszky A. Preparation of purified perikaryal and synaptosomal mitochondrial fractions from relatively small hypothalamic brain samples. MethodsX 2016; 3:417-29. [PMID: 27284533 PMCID: PMC4887559 DOI: 10.1016/j.mex.2016.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/11/2016] [Indexed: 10/25/2022] Open
Abstract
In order to measure the activity of neuronal mitochondria, a representative proof of neuronal processes, physiologically relevant mitochondrial samples need to be gained as simply as possible. Existing methods are, however, either for tissue samples of large size and/or homogenous microstructures only, or are not tested for mitochondrial function measurements. In the present article we describe a gradient fractionation method to isolate viable and well-coupled mitochondria from relatively heterogeneous histological microstructures such as the hypothalamus. With this new method, we are able to isolate a sufficient amount of functional mitochondria for determination of respiratory activity, in a short period of time, using affordable equipment. •Verified by electron microscopy, our method separates highly enriched and well-preserved perikaryal and synaptosomal mitochondria. Both fractions contain minimal cell debris and no myelin. Respiratory measurements (carried out by Clark-type electrode) confirmed undisturbed mitochondrial function providing well-evaluable records. The demonstrated protocol yields highly viable mitochondrial subfractions within 3 h from small brain areas for high-precision examinations. Using this procedure, brain regions with relatively heterogeneous histological microstructure (hypothalamus) can also be efficiently sampled.•Up to our present knowledge, our method is the shortest available procedure with the lowest sample size to gain debris-free, fully-viable mitochondria.
Collapse
Affiliation(s)
- David S Kiss
- Department of Physiology and Biochemistry, Szent Istvan University Faculty of Veterinary Sciences, Budapest, Hungary
| | - Istvan Toth
- Department of Physiology and Biochemistry, Szent Istvan University Faculty of Veterinary Sciences, Budapest, Hungary
| | - Gergely Jocsak
- Department of Physiology and Biochemistry, Szent Istvan University Faculty of Veterinary Sciences, Budapest, Hungary
| | - Agnes Sterczer
- Department and Clinic of Internal Medicine, Szent Istvan University Faculty of Veterinary Sciences, Budapest, Hungary
| | - Tibor Bartha
- Department of Physiology and Biochemistry, Szent Istvan University Faculty of Veterinary Sciences, Budapest, Hungary
| | - Laszlo V Frenyo
- Department of Physiology and Biochemistry, Szent Istvan University Faculty of Veterinary Sciences, Budapest, Hungary
| | - Attila Zsarnovszky
- Division of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Animal Physiology and Animal Health, Szent Istvan University Faculty of Agricultural and Environmental Sciences, Godollo, Hungary
| |
Collapse
|
37
|
Teodoro JS, Silva R, Varela AT, Duarte FV, Rolo AP, Hussain S, Palmeira CM. Low-dose, subchronic exposure to silver nanoparticles causes mitochondrial alterations in Sprague-Dawley rats. Nanomedicine (Lond) 2016; 11:1359-75. [PMID: 27171910 DOI: 10.2217/nnm-2016-0049] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Nanoparticles (NPs) have increasingly been studied due to their probable harmful effects to both humans and the environment. However, despite several indications of possible harmful effects, no long-term studies using a low dose of silver nanoparticles (AgNP) have been conducted in vivo. RESULTS Our data demonstrate that the prolonged exposure to a very low dose of AgNP was sufficient to cause alterations in hepatic mitochondrial function. Mitochondrial function compromised by AgNPs is recovered by pretreatment with the antioxidant N-acetylcysteine, which highlights the crucial role of oxidative stress in AgNPs' toxicity. CONCLUSION Our data show for the first time that even a very low dose of AgNP can cause harmful effects on mitochondrial function, thus compromising the normal function of the organ.
Collapse
Affiliation(s)
- João Soeiro Teodoro
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal.,Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Rui Silva
- Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Ana Teresa Varela
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal.,Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Filipe Valente Duarte
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal.,Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Anabela Pinto Rolo
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal.,Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Saber Hussain
- 711th HPW/RHDJ, Molecular Bioeffects Branch, Bioeffects Division, Human Effectiveness Directorate, Air Force Research Laboratory, Wright Patterson AFB, Dayton, OH 45433, USA
| | - Carlos Marques Palmeira
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal.,Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| |
Collapse
|
38
|
Demarest TG, Schuh RA, Waddell J, McKenna MC, Fiskum G. Sex-dependent mitochondrial respiratory impairment and oxidative stress in a rat model of neonatal hypoxic-ischemic encephalopathy. J Neurochem 2016; 137:714-29. [PMID: 27197831 DOI: 10.1111/jnc.13590] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/12/2016] [Accepted: 02/17/2016] [Indexed: 01/09/2023]
Abstract
Increased male susceptibility to long-term cognitive deficits is well described in clinical and experimental studies of neonatal hypoxic-ischemic encephalopathy. While cell death signaling pathways are known to be sexually dimorphic, a sex-dependent pathophysiological mechanism preceding the majority of secondary cell death has yet to be described. Mitochondrial dysfunction contributes to cell death following cerebral hypoxic-ischemia (HI). Several lines of evidence suggest that there are sex differences in the mitochondrial metabolism of adult mammals. Therefore, this study tested the hypothesis that brain mitochondrial respiratory impairment and associated oxidative stress is more severe in males than females following HI. Maximal brain mitochondrial respiration during oxidative phosphorylation was two-fold more impaired in males following HI. The endogenous antioxidant glutathione was 30% higher in the brain of sham females compared to males. Females also exhibited increased glutathione peroxidase (GPx) activity following HI injury. Conversely, males displayed a reduction in mitochondrial GPx4 protein levels and mitochondrial GPx activity. Moreover, a 3-4-fold increase in oxidative protein carbonylation was observed in the cortex, perirhinal cortex, and hippocampus of injured males, but not females. These data provide the first evidence for sex-dependent mitochondrial respiratory dysfunction and oxidative damage, which may contribute to the relative male susceptibility to adverse long-term outcomes following HI. Lower basal GSH levels, lower post-hypoxic mitochondrial glutathione peroxidase (mtGPx) activity, and mitochondrial glutathione peroxidase 4 (mtGPx4) protein levels may contribute to the susceptibility of the male brain to oxidative damage and mitochondrial dysfunction following neonatal hypoxic-ischemia (HI). Treatment of male pups with acetyl-L-carnitine (ALCAR) protects against the loss of mtGPx activity, mtGPx4 protein, and increases in protein carbonylation after HI. These findings provide novel insight into the pathophysiology of sexually dimorphic outcomes following HI.
Collapse
Affiliation(s)
- Tyler G Demarest
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rosemary A Schuh
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mary C McKenna
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gary Fiskum
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, Maryland, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
39
|
Effect of DSPE-PEG on compound action potential, injury potential and ion concentration following compression in ex vivo spinal cord. Neurosci Lett 2016; 620:50-6. [DOI: 10.1016/j.neulet.2016.03.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 03/01/2016] [Accepted: 03/24/2016] [Indexed: 01/20/2023]
|
40
|
Novgorodov SA, Riley CL, Keffler JA, Yu J, Kindy MS, Macklin WB, Lombard DB, Gudz TI. SIRT3 Deacetylates Ceramide Synthases: IMPLICATIONS FOR MITOCHONDRIAL DYSFUNCTION AND BRAIN INJURY. J Biol Chem 2015; 291:1957-1973. [PMID: 26620563 DOI: 10.1074/jbc.m115.668228] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 12/11/2022] Open
Abstract
Experimental evidence supports the role of mitochondrial ceramide accumulation as a cause of mitochondrial dysfunction and brain injury after stroke. Herein, we report that SIRT3 regulates mitochondrial ceramide biosynthesis via deacetylation of ceramide synthase (CerS) 1, 2, and 6. Reciprocal immunoprecipitation experiments revealed that CerS1, CerS2, and CerS6, but not CerS4, are associated with SIRT3 in cerebral mitochondria. Furthermore, CerS1, -2, and -6 are hyperacetylated in the mitochondria of SIRT3-null mice, and SIRT3 directly deacetylates the ceramide synthases in a NAD(+)-dependent manner that increases enzyme activity. Investigation of the SIRT3 role in mitochondrial response to brain ischemia/reperfusion (IR) showed that SIRT3-mediated deacetylation of ceramide synthases increased enzyme activity and ceramide accumulation after IR. Functional studies demonstrated that absence of SIRT3 rescued the IR-induced blockade of the electron transport chain at the level of complex III, attenuated mitochondrial outer membrane permeabilization, and decreased reactive oxygen species generation and protein carbonyls in mitochondria. Importantly, Sirt3 gene ablation reduced the brain injury after IR. These data support the hypothesis that IR triggers SIRT3-dependent deacetylation of ceramide synthases and the elevation of ceramide, which could inhibit complex III, leading to increased reactive oxygen species generation and brain injury. The results of these studies highlight a novel mechanism of SIRT3 involvement in modulating mitochondrial ceramide biosynthesis and suggest an important role of SIRT3 in mitochondrial dysfunction and brain injury after experimental stroke.
Collapse
Affiliation(s)
- Sergei A Novgorodov
- the Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Christopher L Riley
- From the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| | - Jarryd A Keffler
- the Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jin Yu
- the Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Mark S Kindy
- From the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| | - Wendy B Macklin
- the Department of Cell and Developmental Biology, University of Colorado, Aurora, Colorado 80045, and
| | - David B Lombard
- the Department of Pathology and Institute of Gerontology, University of Michigan, Ann Arbor, Michigan 48109
| | - Tatyana I Gudz
- the Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425,; From the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401,.
| |
Collapse
|
41
|
Schmidt-Kastner R. Genomic approach to selective vulnerability of the hippocampus in brain ischemia–hypoxia. Neuroscience 2015; 309:259-79. [DOI: 10.1016/j.neuroscience.2015.08.034] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 01/06/2023]
|
42
|
Normoyle KP, Kim M, Farahvar A, Llano D, Jackson K, Wang H. The emerging neuroprotective role of mitochondrial uncoupling protein-2 in traumatic brain injury. Transl Neurosci 2015; 6:179-186. [PMID: 28123803 PMCID: PMC4936626 DOI: 10.1515/tnsci-2015-0019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 07/20/2015] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a multifaceted disease with intrinsically complex heterogeneity and remains a significant clinical challenge to manage. TBI model systems have demonstrated many mechanisms that contribute to brain parenchymal cell death, including glutamate and calcium toxicity, oxidative stress, inflammation, and mitochondrial dysfunction. Mitochondria are critically regulated by uncoupling proteins (UCP), which allow protons to leak back into the matrix and thus reduce the mitochondrial membrane potential by dissipating the proton motive force. This uncoupling of oxidative phosphorylation from adenosine triphosphate (ATP) synthesis is potentially critical for protection against cellular injury as a result of TBI and stroke. A greater understanding of the underlying mechanism or mechanisms by which uncoupling protein-2 (UCP2) functions to maintain or optimize mitochondrial function, and the conditions which precipitate the failure of these mechanisms, would inform future research and treatment strategies. We posit that UCP2-mediated function underlies the physiological response to neuronal stress associated with traumatic and ischemic injury and that clinical development of UCP2-targeted treatment would significantly impact these patient populations. With a focus on clinical relevance in TBI, we synthesize current knowledge concerning UCP2 and its potential neuroprotective role and apply this body of knowledge to current and potential treatment modalities.
Collapse
Affiliation(s)
- Kieran P Normoyle
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Miri Kim
- College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Arash Farahvar
- Department of Neurosurgery, Carle Foundation Hospital, Urbana, IL, USA
| | - Daniel Llano
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Neurology, Carle Foundation Hospital, Urbana, IL, USA; The Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Kevin Jackson
- The Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Thermal Neuroscience Laboratory (TNL), Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Huan Wang
- Department of Neurology, Carle Foundation Hospital, Urbana, IL, USA; Thermal Neuroscience Laboratory (TNL), Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
43
|
Rakhunde PB, Saher S, Ali SA. Neuroprotective effect of Feronia limonia on ischemia reperfusion induced brain injury in rats. Indian J Pharmacol 2015; 46:617-21. [PMID: 25538333 PMCID: PMC4264077 DOI: 10.4103/0253-7613.144920] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/08/2014] [Accepted: 10/13/2014] [Indexed: 11/08/2022] Open
Abstract
Objectives: Brain stroke is a leading cause of death without effective treatment. Feronia limonia have potent antioxidant activity and can be proved as neuroprotective against ischemia-reperfusion induced brain injury. Materials and Methods: We studied the effect of methanolic extract of F. limonia fruit (250 mg/kg, 500 mg/kg body weight, p.o.) and Vitamin E as reference standard drug on 30 min induced ischemia, followed by reperfusion by testing the neurobehavioral tests such as neurodeficit score, rota rod test, hanging wire test, beam walk test and elevated plus maze. The biochemical parameters, which were measured in animals brain were catalase, superoxide dismutase (SOD), malondialdehyde and nitric oxide in control and treated rats. Results: The methanolic extract of F. limonia fruit (250 mg/kg, 500 mg/kg body weight, p.o.) treated groups showed a statistically significant improvement in the neurobehavioral parameters such as motor performance (neurological status, significant increase in grasping ability, forelimb strength improvement in balance and co-ordination). The biochemical parameters in the brains of rats showed a significant reduction in the total nitrite (P < 0.01) and lipid peroxidation (P < 0.01), also a significant enhanced activity of enzymatic antioxidants such as catalase (P < 0.01) and SOD (P < 0.05). Conclusion: These observations suggest the neuroprotective and antioxidant activity of F. limonia and Vitamin E on ischemia reperfusion induced brain injury and may require further evaluation.
Collapse
Affiliation(s)
- Purushottam B Rakhunde
- Department of Pharmacology, Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, Maharashtra, India
| | - Sana Saher
- Department of Pharmacology, Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, Maharashtra, India
| | - Syed Ayaz Ali
- Department of Pharmacology, Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, Maharashtra, India
| |
Collapse
|
44
|
Erdoğan H, Tunçdemir M, Kelten B, Akdemir O, Karaoğlan A, Taşdemiroğlu E. The Effects of Difumarate Salt S-15176 after Spinal Cord Injury in Rats. J Korean Neurosurg Soc 2015; 57:445-54. [PMID: 26180614 PMCID: PMC4502243 DOI: 10.3340/jkns.2015.57.6.445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 03/22/2015] [Accepted: 04/09/2015] [Indexed: 01/07/2023] Open
Abstract
Objective In the present study we analyzed neuroprotective and antiapoptotic effect of the difumarate salt S-15176, as an anti-ischemic, an antioxidant and a stabilizer of mitochondrial membrane in secondary damage following spinal cord injury (SCI) in a rat model. Methods Three groups were performed with 30 Wistar rats; control (1), trauma (2), and a trauma+S-15176 (10 mg/kg i.p., dimethyl sulfoxide) treatment (3). SCI was performed at the thoracic level using the weight-drop technique. Spinal cord tissues were collected following intracardiac perfusion in 3rd and 7th days of posttrauma. Hematoxylin and eosin staining for histopatology, terminal deoxynucleotidyl transferase dUTP nick end labeling assay for apoptotic cells and immunohistochemistry for proapoptotic cytochrome-c, Bax and caspase 9 were performed to all groups. Functional recovery test were applied to each group in 3rd and 7th days following SCI. Results In trauma group, edematous regions, diffuse hemorrhage, necrosis, leukocyte infiltration and severe degeneration in motor neurons were observed prominently in gray matter. The number of apoptotic cells was significantly higher (p<0.05) than control group. In the S-15176-treated groups, apoptotic cell number in 3rd and 7th days (p<0.001), also cytochrome-c (p<0.001), Bax (p<0.001) and caspase 9 immunoreactive cells (p<0.001) were significantly decreased in number compared to trauma groups. Hemorrhage and edema in the focal areas were also noticed in gray matter of treatment groups. Results of the locomotor test were significantly increased in treatment group (p<0.05) when compared to trauma groups. Conclusion We suggest that difumarate salt S-15176 prevents mitochondrial pathways of apoptosis and protects spinal cord from secondary injury and helps to preserve motor function following SCI in rats.
Collapse
Affiliation(s)
- Hakan Erdoğan
- Department of Neurosurgery, Faculty of Medicine, Maltepe University, Istanbul, Turkey
| | - Matem Tunçdemir
- Medical Biology Department, Cerrahpasa Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Bilal Kelten
- Department of Neurosurgery, Faculty of Medicine, Maltepe University, Istanbul, Turkey
| | - Osman Akdemir
- Department of Neurosurgery Taksim Education and Research Hospital, Istanbul, Turkey
| | - Alper Karaoğlan
- Department of Neurosurgery, Faculty of Medicine, Maltepe University, Istanbul, Turkey
| | | |
Collapse
|
45
|
Carvalho C, Santos MS, Oliveira CR, Moreira PI. Alzheimer's disease and type 2 diabetes-related alterations in brain mitochondria, autophagy and synaptic markers. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1665-75. [PMID: 25960150 DOI: 10.1016/j.bbadis.2015.05.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 04/13/2015] [Accepted: 05/02/2015] [Indexed: 01/01/2023]
Abstract
We aimed to investigate mitochondrial function, biogenesis and autophagy in the brain of type 2 diabetes (T2D) and Alzheimer's disease (AD) mice. Isolated brain mitochondria and homogenates from cerebral cortex and hippocampus of wild-type (WT), triple transgenic AD (3xTg-AD) and T2D mice were used to evaluate mitochondrial functional parameters and protein levels of mitochondrial biogenesis, autophagy and synaptic integrity markers, respectively. A significant decrease in mitochondrial respiration, membrane potential and energy levels was observed in T2D and 3xTg-AD mice. Also, a significant decrease in the levels of autophagy-related protein 7 (ATG7) and glycosylated lysosomal membrane protein 1 (LAMP1) was observed in cerebral cortex and hippocampus of T2D and 3xTg-AD mice. Moreover, both brain regions of 3xTg-AD mice present lower levels of nuclear respiratory factor (NRF) 1 while the levels of NRF2 are lower in both brain regions of T2D and 3xTg-AD mice. A decrease in mitochondrial encoded, nicotinamide adenine dinucleotide dehydrogenase subunit 1 (ND1) was also observed in T2D and 3xTg-AD mice although only statistically significant in T2D cortex. Furthermore, a decrease in the levels of postsynaptic density protein 95 (PSD95) in the cerebral cortex of 3xTg-AD mice and in hippocampus of T2D and 3xTg-AD mice and a decrease in the levels of synaptosomal-associated protein 25 (SNAP 25) in the hippocampus of T2D and 3xTg-AD mice were observed suggesting synaptic integrity loss. These results support the idea that alterations in mitochondrial function, biogenesis and autophagy cause synaptic damage in AD and T2D.
Collapse
Affiliation(s)
- Cristina Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal.
| | - Maria S Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Catarina R Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Laboratory of Biochemistry, Faculty of Medicine, University of Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
46
|
Arsalandeh F, Ahmadian S, Foolad F, Khodagholi F, Farimani MM, Shaerzadeh F. Beneficial Effect of Flavone Derivatives on Aβ-Induced Memory Deficit Is Mediated by Peroxisome Proliferator-Activated Receptor γ Coactivator 1α: A Comparative Study. Int J Toxicol 2015; 34:274-83. [PMID: 25972379 DOI: 10.1177/1091581815584165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
In the present study, the neuroprotective effect of 5-hydroxy-6,7,4'-trimethoxyflavone (flavone 1), a natural flavone, was investigated in comparison with another flavone, 5,7,4'-trihydroxyflavone (flavone 2) on the hippocampus of amyloid beta (Aβ)-injected rats. Rats were treated with the 2 flavones (1 mg/kg/d) for 1 week before Aβ injection. Seven days after Aβ administration, memory function of rats was assessed in a passive avoidance test (PAT). Changes in the levels of mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor γ coactivator 1 α (PGC-1α), phospho-adenosine monophosphate (AMP)-activated protein kinase (pAMPK), AMPK, phospho-cAMP-responsive element-binding protein (CREB), CREB, and nuclear respiratory factor 1 (NRF-1) proteins were determined by Western blot analysis. Our results showed an improvement in memory in rats pretreated with flavonoids. At the molecular level, phosphorylation of CREB, known as the master modulator of memory processes, increased. On the other hand, the level of mitochondrial biogenesis factors, PGC-1α and its downstream molecules NRF-1 and TFAM significantly increased by dietary administration of 2 flavones. In addition, flavone 1 and flavone 2 prevented mitochondrial swelling and mitochondrial membrane potential reduction. Our results provided evidence that flavone 1 is more effective than flavone 2 presumably due to its O-methylated groups. In conclusion, it seems that in addition to classical antioxidant effect, flavones exert part of their protective effects through mitochondrial biogenesis.
Collapse
Affiliation(s)
- Farshad Arsalandeh
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Shahin Ahmadian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Forough Foolad
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi M Farimani
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G. C., Evin, Tehran, Iran
| | - Fatemeh Shaerzadeh
- Department of Physiology, Faculty of Medicine, Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Bedside diagnosis of mitochondrial dysfunction after malignant middle cerebral artery infarction. Neurocrit Care 2015; 21:35-42. [PMID: 23860668 DOI: 10.1007/s12028-013-9875-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND The study explores whether the cerebral biochemical pattern in patients treated with hemicraniectomy after large middle cerebral artery infarcts reflects ongoing ischemia or non-ischemic mitochondrial dysfunction. METHODS The study includes 44 patients treated with decompressive hemicraniectomy (DCH) due to malignant middle cerebral artery infarctions. Chemical variables related to energy metabolism obtained by microdialysis were analyzed in the infarcted tissue and in the contralateral hemisphere from the time of DCH until 96 h after DCH. RESULTS Reperfusion of the infarcted tissue was documented in a previous report. Cerebral lactate/pyruvate ratio (L/P) and lactate were significantly elevated in the infarcted tissue compared to the non-infarcted hemisphere (p < 0.05). From 12 to 96 h after DCH the pyruvate level was significantly higher in the infarcted tissue than in the non-infarcted hemisphere (p < 0.05). CONCLUSION After a prolonged period of ischemia and subsequent reperfusion, cerebral tissue shows signs of protracted mitochondrial dysfunction, characterized by a marked increase in cerebral lactate level with a normal or increased cerebral pyruvate level resulting in an increased LP-ratio. This biochemical pattern contrasts to cerebral ischemia, which is characterized by a marked decrease in cerebral pyruvate. The study supports the hypothesis that it is possible to diagnose cerebral mitochondrial dysfunction and to separate it from cerebral ischemia by microdialysis and bed-side biochemical analysis.
Collapse
|
48
|
Kenborg L, Rugbjerg K, Lee PC, Ravnskjær L, Christensen J, Ritz B, Lassen CF. Head injury and risk for Parkinson disease: results from a Danish case-control study. Neurology 2015; 84:1098-103. [PMID: 25681453 PMCID: PMC4371406 DOI: 10.1212/wnl.0000000000001362] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/29/2014] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE To examine the association between head injuries throughout life and the risk for Parkinson disease (PD) in an interview-based case-control study. METHODS We identified 1,705 patients diagnosed with PD at 10 neurologic centers in Denmark in 1996-2009 and verified their diagnoses in medical records. Patients were matched to 1,785 controls randomly selected from the Danish Central Population Register on sex and year of birth. Odds ratios (ORs) and 95% confidence intervals (CIs) were estimated using unconditional logistic regression. RESULTS We observed no association between any head injury before first cardinal symptom and PD (OR 1.02; 95% CI 0.88, 1.19). Examination of number of head injuries (1: OR 1.02; 95% CI 0.87, 1.20; ≥2: OR 1.03; 95% CI 0.72, 1.47) or hospitalization for a head injury (OR 0.89; 95% CI 0.70, 1.12) did not show an association with PD. For 954 study subjects with at least one head injury, there was no evidence of an association between loss of consciousness (OR 0.89; 95% CI 0.67, 1.17), duration of loss of consciousness (≤1 minute: OR 0.93; 95% CI 0.58, 1.49; 1-5 minutes: OR 0.74; 95% CI 0.51, 1.08; ≥5 minutes: OR 0.81; 95% CI 0.53, 1.24), or amnesia (OR 1.31; 95% CI 0.88, 1.95) and risk for PD. Application of a lag time of 10 years between head injury and first cardinal symptom resulted in similar risk estimates. CONCLUSIONS The results do not support the hypothesis that head injury increases the risk for PD.
Collapse
Affiliation(s)
- Line Kenborg
- From the Danish Cancer Society Research Center (L.K., K.R., L.R., J.C., C.F.L.), Danish Cancer Society, Copenhagen, Denmark; the Department of Epidemiology, Fielding School of Public Health (P.-C.L., B.R.), and the Department of Neurology, School of Medicine (B.R.), University of California, Los Angeles; the Department of Health Care Management (P.-C.L.), College of Healthcare Administration and Management, National Taipei University of Nursing Health Sciences, Taipei, Taiwan; and the Department of Occupational and Environmental Medicine (C.F.L.), Bispebjerg Hospital, Copenhagen, Denmark.
| | - Kathrine Rugbjerg
- From the Danish Cancer Society Research Center (L.K., K.R., L.R., J.C., C.F.L.), Danish Cancer Society, Copenhagen, Denmark; the Department of Epidemiology, Fielding School of Public Health (P.-C.L., B.R.), and the Department of Neurology, School of Medicine (B.R.), University of California, Los Angeles; the Department of Health Care Management (P.-C.L.), College of Healthcare Administration and Management, National Taipei University of Nursing Health Sciences, Taipei, Taiwan; and the Department of Occupational and Environmental Medicine (C.F.L.), Bispebjerg Hospital, Copenhagen, Denmark
| | - Pei-Chen Lee
- From the Danish Cancer Society Research Center (L.K., K.R., L.R., J.C., C.F.L.), Danish Cancer Society, Copenhagen, Denmark; the Department of Epidemiology, Fielding School of Public Health (P.-C.L., B.R.), and the Department of Neurology, School of Medicine (B.R.), University of California, Los Angeles; the Department of Health Care Management (P.-C.L.), College of Healthcare Administration and Management, National Taipei University of Nursing Health Sciences, Taipei, Taiwan; and the Department of Occupational and Environmental Medicine (C.F.L.), Bispebjerg Hospital, Copenhagen, Denmark
| | - Line Ravnskjær
- From the Danish Cancer Society Research Center (L.K., K.R., L.R., J.C., C.F.L.), Danish Cancer Society, Copenhagen, Denmark; the Department of Epidemiology, Fielding School of Public Health (P.-C.L., B.R.), and the Department of Neurology, School of Medicine (B.R.), University of California, Los Angeles; the Department of Health Care Management (P.-C.L.), College of Healthcare Administration and Management, National Taipei University of Nursing Health Sciences, Taipei, Taiwan; and the Department of Occupational and Environmental Medicine (C.F.L.), Bispebjerg Hospital, Copenhagen, Denmark
| | - Jane Christensen
- From the Danish Cancer Society Research Center (L.K., K.R., L.R., J.C., C.F.L.), Danish Cancer Society, Copenhagen, Denmark; the Department of Epidemiology, Fielding School of Public Health (P.-C.L., B.R.), and the Department of Neurology, School of Medicine (B.R.), University of California, Los Angeles; the Department of Health Care Management (P.-C.L.), College of Healthcare Administration and Management, National Taipei University of Nursing Health Sciences, Taipei, Taiwan; and the Department of Occupational and Environmental Medicine (C.F.L.), Bispebjerg Hospital, Copenhagen, Denmark
| | - Beate Ritz
- From the Danish Cancer Society Research Center (L.K., K.R., L.R., J.C., C.F.L.), Danish Cancer Society, Copenhagen, Denmark; the Department of Epidemiology, Fielding School of Public Health (P.-C.L., B.R.), and the Department of Neurology, School of Medicine (B.R.), University of California, Los Angeles; the Department of Health Care Management (P.-C.L.), College of Healthcare Administration and Management, National Taipei University of Nursing Health Sciences, Taipei, Taiwan; and the Department of Occupational and Environmental Medicine (C.F.L.), Bispebjerg Hospital, Copenhagen, Denmark
| | - Christina F Lassen
- From the Danish Cancer Society Research Center (L.K., K.R., L.R., J.C., C.F.L.), Danish Cancer Society, Copenhagen, Denmark; the Department of Epidemiology, Fielding School of Public Health (P.-C.L., B.R.), and the Department of Neurology, School of Medicine (B.R.), University of California, Los Angeles; the Department of Health Care Management (P.-C.L.), College of Healthcare Administration and Management, National Taipei University of Nursing Health Sciences, Taipei, Taiwan; and the Department of Occupational and Environmental Medicine (C.F.L.), Bispebjerg Hospital, Copenhagen, Denmark
| |
Collapse
|
49
|
Head trauma in sport and neurodegenerative disease: an issue whose time has come? Neurobiol Aging 2014; 36:1383-9. [PMID: 25725943 DOI: 10.1016/j.neurobiolaging.2014.12.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 12/13/2022]
Abstract
A number of small studies and anecdotal reports have been suggested that sports involving repeated head trauma may have long-term risks of neurodegenerative disease. There are now plausible mechanisms for these effects, and a recognition that these problems do not just occur in former boxers, but in a variety of sports involving repeated concussions, and possibly also in sports in which low-level head trauma is common. These neurodegenerative effects potentially include increased risks of impaired cognitive function and dementia, Parkinson's disease, and amyotrophic lateral sclerosis. Many would argue for taking a precautionary approach and immediately banning or restricting sports such as boxing. However, there are important public health issues in terms of how wide the net should be cast in terms of other sports, and what remedial measures could be taken? This in turn requires a major research effort involving both clinical and basic research to understand the underlying mechanisms, leading from head trauma to neurodegenerative disease and epidemiologic studies to assess the long-term consequences.
Collapse
|
50
|
Shoshan-Barmatz V, Ben-Hail D, Admoni L, Krelin Y, Tripathi SS. The mitochondrial voltage-dependent anion channel 1 in tumor cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2547-75. [PMID: 25448878 DOI: 10.1016/j.bbamem.2014.10.040] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 02/06/2023]
Abstract
VDAC1 is found at the crossroads of metabolic and survival pathways. VDAC1 controls metabolic cross-talk between mitochondria and the rest of the cell by allowing the influx and efflux of metabolites, ions, nucleotides, Ca2+ and more. The location of VDAC1 at the outer mitochondrial membrane also enables its interaction with proteins that mediate and regulate the integration of mitochondrial functions with cellular activities. As a transporter of metabolites, VDAC1 contributes to the metabolic phenotype of cancer cells. Indeed, this protein is over-expressed in many cancer types, and silencing of VDAC1 expression induces an inhibition of tumor development. At the same time, along with regulating cellular energy production and metabolism, VDAC1 is involved in the process of mitochondria-mediated apoptosis by mediating the release of apoptotic proteins and interacting with anti-apoptotic proteins. The engagement of VDAC1 in the release of apoptotic proteins located in the inter-membranal space involves VDAC1 oligomerization that mediates the release of cytochrome c and AIF to the cytosol, subsequently leading to apoptotic cell death. Apoptosis can also be regulated by VDAC1, serving as an anchor point for mitochondria-interacting proteins, such as hexokinase (HK), Bcl2 and Bcl-xL, some of which are also highly expressed in many cancers. By binding to VDAC1, HK provides both a metabolic benefit and apoptosis-suppressive capacity that offer the cell a proliferative advantage and increase its resistance to chemotherapy. Thus, these and other functions point to VDAC1 as an excellent target for impairing the re-programed metabolism of cancer cells and their ability to evade apoptosis. Here, we review current evidence pointing to the function of VDAC1 in cell life and death, and highlight these functions in relation to both cancer development and therapy. In addressing the recently solved 3D structures of VDAC1, this review will point to structure-function relationships of VDAC as critical for deciphering how this channel can perform such a variety of roles, all of which are important for cell life and death. Finally, this review will also provide insight into VDAC function in Ca2+ homeostasis, protection against oxidative stress, regulation of apoptosis and involvement in several diseases, as well as its role in the action of different drugs. We will discuss the use of VDAC1-based strategies to attack the altered metabolism and apoptosis of cancer cells. These strategies include specific siRNA able to impair energy and metabolic homeostasis, leading to arrested cancer cell growth and tumor development, as well VDAC1-based peptides that interact with anti-apoptotic proteins to induce apoptosis, thereby overcoming the resistance of cancer cell to chemotherapy. Finally, small molecules targeting VDAC1 can induce apoptosis. VDAC1 can thus be considered as standing at the crossroads between mitochondrial metabolite transport and apoptosis and hence represents an emerging cancer drug target. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | - Danya Ben-Hail
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Lee Admoni
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Yakov Krelin
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Shambhoo Sharan Tripathi
- Department of Life Sciences, and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|