1
|
Zeng Q, Li L, Chang T, Sun Y, Zheng B, Xue L, Liu C, Li X, Huang R, Gu J, An Z, Yao H, Zhou D, Fan J, Dai Y. Phosphorylation of POU3F3 Mediated Nuclear Translocation Promotes Proliferation in Non-Small Cell Lung Cancer through Accelerating ATP5PF Transcription and ATP Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411503. [PMID: 39932442 PMCID: PMC11967767 DOI: 10.1002/advs.202411503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/09/2025] [Indexed: 04/05/2025]
Abstract
Targeting oxidative phosphorylation (OXPHOS) through inhibiting the electron transport chain (ETC) has shown promising pre-clinical efficacy in cancer therapy. Although aerobic glycolysis is a hallmark of cancer, emerging evidence suggest OXPHOS is frequently enhanced, providing metabolic advantages for cell proliferation, metastasis, and drug resistance in a variety of aggressive cancer types including non-small cell lung cancer (NSCLC), yet the underlying molecular mechanisms remain elusive. Here it is reported that POU-domain containing family protein POU3F3 is translocated into the nuclei of NSCLC cell lines harboring mutant RAS, where it activates transcription of ATP5PF, an essential component of mitochondrial ATP synthase and consequent ATP production, leading to enhanced NSCLC proliferation and migration. Moreover, it is further found out that ERK1 phosphorylates POU3F3 at the S393 site in the cytoplasm and promotes the nuclear translocation of POU3F3 via receptor importin β1 in RAS mutant NSCLC cells. Mechanistically, RNA sequencing analysis combined with chromatin immunoprecipitation (ChIP) assay revealed that POU3F3 binds to the promoter of ATP5PF, leading to enhanced ATP5PF transcription and ATP production. Together, this study uncovers a novel RAS-POU3F3-ATP5PF axis in facilitating NSCLC progression, providing a new perspective on the understanding of molecular mechanisms for NSCLC progression.
Collapse
Affiliation(s)
- Qi‐Gang Zeng
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Le Li
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Tao Chang
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Yong Sun
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Bin Zheng
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Ling‐Na Xue
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Chao‐Ling Liu
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| | - Xia‐Qing Li
- Institute of Nephrology and Blood PurificationThe First Affiliated HospitalJinan UniversityGuangdong510632China
- Nephrology departmentThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityGuangdong517000China
| | - Ruo‐Tong Huang
- Department of Metabolism, Digestion, and ReproductionFaculty of MedicineImperial College LondonLondonW12 0NNUK
| | - Jia‐Xin Gu
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Zhao‐Rong An
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Hao‐Tao Yao
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
| | - Dan‐Yang Zhou
- Department of RespiratoryNanjing First HospitalNanjing Medical UniversityJiangsu210012China
| | - Jun Fan
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
- Department of Medical Biochemistry and Molecular BiologySchool of MedicineJinan UniversityGuangdong510632China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University)Ministry of EducationGuangdong510632China
| | - Yong Dai
- Nanhai hospital of Traditional Chinese MedicineJinan UniversityGuangdong528200China
| |
Collapse
|
2
|
Musyaju S, Modi HR, Shear DA, Scultetus AH, Pandya JD. Time Course of Mitochondrial Antioxidant Markers in a Preclinical Model of Severe Penetrating Traumatic Brain Injury. Int J Mol Sci 2025; 26:906. [PMID: 39940675 PMCID: PMC11816813 DOI: 10.3390/ijms26030906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
Traumatic brain injury (TBI) results from external mechanical forces exerted on the brain, triggering secondary injuries due to cellular excitotoxicity. A key indicator of damage is mitochondrial dysfunction, which is associated with elevated free radicals and disrupted redox balance following TBI. However, the temporal changes in mitochondrial redox homeostasis after penetrating TBI (PTBI) have not been thoroughly examined. This study aimed to investigate redox alterations from 30 min to two-weeks post-injury in adult male Sprague Dawley rats that experienced either PTBI or a Sham craniectomy. Redox parameters were measured at several points: 30 min, 3 h, 6 h, 24 h, 3 d, 7 d, and 14 d post-injury. Mitochondrial samples from the injury core and perilesional areas exhibited significant elevations in protein modifications including 3-nitrotyrosine (3-NT) and protein carbonyl (PC) adducts (14-53%, vs. Sham). In parallel, antioxidants such as glutathione, NADPH, peroxiredoxin-3 (PRX-3), thioredoxin-2 (TRX-2), and superoxide dismutase 2 (SOD2) were significantly depleted (20-80%, vs. Sham). In contrast, catalase (CAT) expression showed a significant increase (45-75%, vs. Sham). These findings indicate a notable imbalance in redox parameters over the two-week post-PTBI period suggesting that the therapeutic window to employ antioxidant therapy extends well beyond 24 h post-TBI.
Collapse
Affiliation(s)
| | | | | | | | - Jignesh D. Pandya
- Brain Trauma Neuroprotection (BTN) Branch, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD 20910, USA
| |
Collapse
|
3
|
Kapapa T, Wernheimer V, Hoffmann A, Merz T, Zink F, Wolfschmitt EM, McCook O, Vogt J, Wepler M, Messerer DAC, Hartmann C, Scheuerle A, Mathieu R, Mayer S, Gröger M, Denoix N, Clazia E, Radermacher P, Röhrer S, Datzmann T. Unravelling Secondary Brain Injury: Insights from a Human-Sized Porcine Model of Acute Subdural Haematoma. Cells 2024; 14:17. [PMID: 39791718 PMCID: PMC11720468 DOI: 10.3390/cells14010017] [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: 10/06/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
Traumatic brain injury (TBI) remains one of the leading causes of death. Because of the individual nature of the trauma (brain, circumstances and forces), humans experience individual TBIs. This makes it difficult to generalise therapies. Clinical management issues such as whether intracranial pressure (ICP), cerebral perfusion pressure (CPP) or decompressive craniectomy improve patient outcome remain partly unanswered. Experimental drug approaches for the treatment of secondary brain injury (SBI) have not found clinical application. The complex, cellular and molecular pathways of SBI remain incompletely understood, and there are insufficient experimental (animal) models that reflect the pathophysiology of human TBI to develop translational therapeutic approaches. Therefore, we investigated different injury patterns after acute subdural hematoma (ASDH) as TBI in a post-hoc approach to assess the impact on SBI in a long-term, human-sized porcine TBI animal model. Post-mortem brain tissue analysis, after ASDH, bilateral ICP, CPP, cerebral oxygenation and temperature monitoring, and biomarker analysis were performed. Extracerebral, intraparenchymal-extraventricular and intraventricular blood, combined with brainstem and basal ganglia injury, influenced the experiment and its outcome. Basal ganglia injury affects the duration of the experiment. Recognition of these different injury patterns is important for translational interpretation of results in this animal model of SBI after TBI.
Collapse
Affiliation(s)
- Thomas Kapapa
- Department of Neurosurgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Vanida Wernheimer
- Department of Neurosurgery, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Andrea Hoffmann
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Tamara Merz
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Fabia Zink
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Eva-Maria Wolfschmitt
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Oscar McCook
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Josef Vogt
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Martin Wepler
- Department of Anaesthesiology, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | | | - Claire Hartmann
- Department of Anaesthesiology, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Angelika Scheuerle
- Section Neuropathology, University Hospital Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - René Mathieu
- Department of Neurosurgery, Military Hospital Ulm, Oberer Eselsberg 40, 89081 Ulm, Germany
| | - Simon Mayer
- Department of Neurosurgery, Military Hospital Ulm, Oberer Eselsberg 40, 89081 Ulm, Germany
| | - Michael Gröger
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Nicole Denoix
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Enrico Clazia
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Peter Radermacher
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Stefan Röhrer
- Department of Neurosurgery, Ostalb-Hospital Aalen, Im Kälblesrain 1, 73430 Aalen, Germany
| | - Thomas Datzmann
- Institute of Anaesthesiologic Pathophysiology and Process Development, University Hospital Ulm, Helmholtzstrasse 8/1, 89081 Ulm, Germany
| |
Collapse
|
4
|
Hu Q, Wu X, Guo C, Wang T, Guo H, Wang J, Wang B, Cui W, Bai H, Zhou J, Li L, Han L, Cao L, Ge S, Gao G, Wang T, Wu Z, Guo W, Qu Y, Feng J, Liu H. Astrocyte-neuron crosstalk through extracellular vesicle-shuttled miRNA-382-5p promotes traumatic brain injury. Exp Mol Med 2024; 56:2642-2658. [PMID: 39617787 DOI: 10.1038/s12276-024-01355-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/26/2024] [Accepted: 08/27/2024] [Indexed: 12/28/2024] Open
Abstract
Although astrocytes undergo functional changes in response to brain injury and may be the driving force of subsequent neuronal death, the underlying mechanisms remain incompletely elucidated. Here, we showed that extracellular vesicle (EV)-shuttled miRNA-382-5p may serve as a biomarker for the severity of traumatic brain injury (TBI), as the circulating EV-miRNA-382-5p level was significantly increased in both human patients and TBI model mice. Mechanistically, astrocyte-derived EVs delivered the shuttled miRNA-382-5p to mediate astrocyte-neuron communication, which promoted neuronal mitochondrial dysfunction by inhibiting the expression of optic atrophy-1 (OPA1). Consistent with these findings, genetic ablation of neuronal OPA1 exacerbated mitochondrial damage and neuronal apoptosis in response to TBI. Moreover, engineered RVG-miRNA-382-5p inhibitor-EVs, which can selectively deliver a miRNA-382-5p inhibitor to neurons, significantly attenuated mitochondrial damage and improved neurological function after TBI. Taken together, our data suggest that EV-shuttled miRNA-382-5p may be a critical mediator of astrocyte-induced neurotoxicity under pathological conditions and that targeting miRNA-382-5p-OPA1 signaling has potential for clinical translation in the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Qing Hu
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Chengxuan Guo
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Tinghao Wang
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Hao Guo
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
- Shaanxi Clinical Research Center for Neurosurgical Diseases, Xi'an, Shaanxi, China
| | - Jin Wang
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
- Department of Neurosurgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Bodong Wang
- Department of Neurosurgery, The 960th Hospital of the PLA Joint Logistics Support Force, Jinan, Shandong, China
| | - Wenxing Cui
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Hao Bai
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Jinpeng Zhou
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
- Shaanxi Clinical Research Center for Neurosurgical Diseases, Xi'an, Shaanxi, China
| | - Leiyang Li
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Liying Han
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Liang Cao
- Department of Traditional Chinese Medicine, Tangdu Hospital, Xi'an, Shaanxi, China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
- Shaanxi Clinical Research Center for Neurosurgical Diseases, Xi'an, Shaanxi, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
- Shaanxi Clinical Research Center for Neurosurgical Diseases, Xi'an, Shaanxi, China
| | - Ting Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhenyong Wu
- University of Chinese Academy of Sciences, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wei Guo
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China
- Shaanxi Clinical Research Center for Neurosurgical Diseases, Xi'an, Shaanxi, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China.
- Shaanxi Clinical Research Center for Neurosurgical Diseases, Xi'an, Shaanxi, China.
| | - Jing Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, Xi'an, Shaanxi, China.
- Shaanxi Clinical Research Center for Neurosurgical Diseases, Xi'an, Shaanxi, China.
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
5
|
Senthil K, Ranganathan A, Piel S, Hefti MM, Reeder RW, Kirschen MP, Starr J, Morton S, Gaudio HA, Slovis JC, Herrmann JR, Berg RA, Kilbaugh TJ, Morgan RW. Elevated serum neurologic biomarker profiles after cardiac arrest in a porcine model. Resusc Plus 2024; 19:100726. [PMID: 39149222 PMCID: PMC11325790 DOI: 10.1016/j.resplu.2024.100726] [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: 04/19/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction Swine exhibit cerebral cortex mitochondrial dysfunction and neuropathologic injury after hypoxic cardiac arrest treated with hemodynamic-directed CPR (HD-CPR) despite normal Cerebral Performance Category scores. We analyzed the temporal evolution of plasma protein biomarkers of brain injury and inflammatory cytokines, as well as cerebral cortical mitochondrial injury and neuropathology for five days following pediatric asphyxia-associated cardiac arrest treated with HD-CPR. Methods One-month-old swine underwent asphyxia associated cardiac arrest, 10-20 min of HD-CPR (goal SBP 90 mmHg, coronary perfusion pressure 20 mmHg), and randomization to post-ROSC survival duration (24, 48, 72, 96, 120 h; n = 3 per group) with standardized post-resuscitation care. Plasma neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and cytokine levels were collected pre-injury and 1, 6, 24, 48, 72, 96, and 120 h post-ROSC. Cerebral cortical tissue was assessed for: mitochondrial respirometry, mass, and dynamic proteins; oxidative injury; and neuropathology. Results Relative to pre-arrest baseline (9.4 pg/ml [6.7-12.6]), plasma NfL was increased at all post-ROSC time points. Each sequential NfL measurement through 48 h was greater than the previous value {1 h (12.7 pg/ml [8.4-14.6], p = 0.01), 6 h (30.9 pg/ml [17.7-44.0], p = 0.0004), 24 h (59.4 pg/ml [50.8-96.1], p = 0.0003) and 48 h (85.7 pg/ml [61.9-118.7], p = 0.046)}. Plasma GFAP, inflammatory cytokines or cerebral cortical tissue measurements were not demonstrably different between time points. Conclusions In a swine model of pediatric cardiac arrest, plasma NfL had an upward trajectory until 48 h post-ROSC after which it remained elevated through five days, suggesting it may be a sensitive marker of neurologic injury following pediatric cardiac arrest.
Collapse
Affiliation(s)
- Kumaran Senthil
- Children's Hospital of Philadelphia and Perelman School of Medicine at University of Pennsylvania, Department of Anesthesiology and Critical Care Medicine, USA
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Abhay Ranganathan
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Sarah Piel
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
- University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Department of Cardiology, Pulmonology and Vascular Medicine, Germany
- University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Cardiovascular Research Institute, Germany
| | | | - Ron W Reeder
- University of Utah, Department of Pediatrics, USA
| | - Matthew P Kirschen
- Children's Hospital of Philadelphia and Perelman School of Medicine at University of Pennsylvania, Department of Anesthesiology and Critical Care Medicine, USA
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Jonathan Starr
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Sarah Morton
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Hunter A Gaudio
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Julia C Slovis
- Children's Hospital of Philadelphia and Perelman School of Medicine at University of Pennsylvania, Department of Anesthesiology and Critical Care Medicine, USA
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Jeremy R Herrmann
- Children's Hospital of Philadelphia and Perelman School of Medicine at University of Pennsylvania, Department of Anesthesiology and Critical Care Medicine, USA
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Robert A Berg
- Children's Hospital of Philadelphia and Perelman School of Medicine at University of Pennsylvania, Department of Anesthesiology and Critical Care Medicine, USA
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Todd J Kilbaugh
- Children's Hospital of Philadelphia and Perelman School of Medicine at University of Pennsylvania, Department of Anesthesiology and Critical Care Medicine, USA
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| | - Ryan W Morgan
- Children's Hospital of Philadelphia and Perelman School of Medicine at University of Pennsylvania, Department of Anesthesiology and Critical Care Medicine, USA
- Children's Hospital of Philadelphia, Resuscitation Science Center, USA
| |
Collapse
|
6
|
Peper CJ, Kilgore MD, Jiang Y, Xiu Y, Xia W, Wang Y, Shi M, Zhou D, Dumont AS, Wang X, Liu N. Tracing the path of disruption: 13C isotope applications in traumatic brain injury-induced metabolic dysfunction. CNS Neurosci Ther 2024; 30:e14693. [PMID: 38544365 PMCID: PMC10973562 DOI: 10.1111/cns.14693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 05/14/2024] Open
Abstract
Cerebral metabolic dysfunction is a critical pathological hallmark observed in the aftermath of traumatic brain injury (TBI), as extensively documented in clinical investigations and experimental models. An in-depth understanding of the bioenergetic disturbances that occur following TBI promises to reveal novel therapeutic targets, paving the way for the timely development of interventions to improve patient outcomes. The 13C isotope tracing technique represents a robust methodological advance, harnessing biochemical quantification to delineate the metabolic trajectories of isotopically labeled substrates. This nuanced approach enables real-time mapping of metabolic fluxes, providing a window into the cellular energetic state and elucidating the perturbations in key metabolic circuits. By applying this sophisticated tool, researchers can dissect the complexities of bioenergetic networks within the central nervous system, offering insights into the metabolic derangements specific to TBI pathology. Embraced by both animal studies and clinical research, 13C isotope tracing has bolstered our understanding of TBI-induced metabolic dysregulation. This review synthesizes current applications of isotope tracing and its transformative potential in evaluating and addressing the metabolic sequelae of TBI.
Collapse
Affiliation(s)
- Charles J. Peper
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Mitchell D. Kilgore
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yinghua Jiang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yuwen Xiu
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Winna Xia
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yingjie Wang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Mengxuan Shi
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Di Zhou
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Aaron S. Dumont
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
- Neuroscience Program, Tulane Brain InstituteTulane UniversityNew OrleansLouisianaUSA
| | - Ning Liu
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
- Neuroscience Program, Tulane Brain InstituteTulane UniversityNew OrleansLouisianaUSA
- Tulane University Translational Sciences InstituteNew OrleansLouisianaUSA
| |
Collapse
|
7
|
Pandya JD, Musyaju S, Modi HR, Okada-Rising SL, Bailey ZS, Scultetus AH, Shear DA. Intranasal delivery of mitochondria targeted neuroprotective compounds for traumatic brain injury: screening based on pharmacological and physiological properties. J Transl Med 2024; 22:167. [PMID: 38365798 PMCID: PMC10874030 DOI: 10.1186/s12967-024-04908-2] [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: 11/21/2023] [Accepted: 01/18/2024] [Indexed: 02/18/2024] Open
Abstract
Targeting drugs to the mitochondrial level shows great promise for acute and chronic treatment of traumatic brain injury (TBI) in both military and civilian sectors. Perhaps the greatest obstacle to the successful delivery of drug therapies is the blood brain barrier (BBB). Intracerebroventricular and intraparenchymal routes may provide effective delivery of small and large molecule therapies for preclinical neuroprotection studies. However, clinically these delivery methods are invasive, and risk inadequate exposure to injured brain regions due to the rapid turnover of cerebral spinal fluid. The direct intranasal drug delivery approach to therapeutics holds great promise for the treatment of central nervous system (CNS) disorders, as this route is non-invasive, bypasses the BBB, enhances the bioavailability, facilitates drug dose reduction, and reduces adverse systemic effects. Using the intranasal method in animal models, researchers have successfully reduced stroke damage, reversed Alzheimer's neurodegeneration, reduced anxiety, improved memory, and delivered neurotrophic factors and neural stem cells to the brain. Based on literature spanning the past several decades, this review aims to highlight the advantages of intranasal administration over conventional routes for TBI, and other CNS disorders. More specifically, we have identified and compiled a list of most relevant mitochondria-targeted neuroprotective compounds for intranasal administration based on their mechanisms of action and pharmacological properties. Further, this review also discusses key considerations when selecting and testing future mitochondria-targeted drugs given intranasally for TBI.
Collapse
Affiliation(s)
- Jignesh D Pandya
- TBI Bioenergetics, Metabolism and Neurotherapeutics Program, Brain Trauma Neuroprotection (BTN) Branch, Center for Military Psychiatry and Neuroscience (CMPN), Walter Reed Army Institute of Research (WRAIR), 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA.
| | - Sudeep Musyaju
- TBI Bioenergetics, Metabolism and Neurotherapeutics Program, Brain Trauma Neuroprotection (BTN) Branch, Center for Military Psychiatry and Neuroscience (CMPN), Walter Reed Army Institute of Research (WRAIR), 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Hiren R Modi
- TBI Bioenergetics, Metabolism and Neurotherapeutics Program, Brain Trauma Neuroprotection (BTN) Branch, Center for Military Psychiatry and Neuroscience (CMPN), Walter Reed Army Institute of Research (WRAIR), 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Starlyn L Okada-Rising
- TBI Bioenergetics, Metabolism and Neurotherapeutics Program, Brain Trauma Neuroprotection (BTN) Branch, Center for Military Psychiatry and Neuroscience (CMPN), Walter Reed Army Institute of Research (WRAIR), 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Zachary S Bailey
- TBI Bioenergetics, Metabolism and Neurotherapeutics Program, Brain Trauma Neuroprotection (BTN) Branch, Center for Military Psychiatry and Neuroscience (CMPN), Walter Reed Army Institute of Research (WRAIR), 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Anke H Scultetus
- TBI Bioenergetics, Metabolism and Neurotherapeutics Program, Brain Trauma Neuroprotection (BTN) Branch, Center for Military Psychiatry and Neuroscience (CMPN), Walter Reed Army Institute of Research (WRAIR), 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Deborah A Shear
- TBI Bioenergetics, Metabolism and Neurotherapeutics Program, Brain Trauma Neuroprotection (BTN) Branch, Center for Military Psychiatry and Neuroscience (CMPN), Walter Reed Army Institute of Research (WRAIR), 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| |
Collapse
|
8
|
Lin X, Zhang H, Chu Y, Zhang Y, Xu C, Xie H, Ruan Q, Lin J, Huang C, Chai D. Honokiol ameliorates angiotensin II-induced cardiac hypertrophy by promoting dissociation of the Nur77-LKB1 complex and activating the AMPK pathway. J Cell Mol Med 2024; 28:e18028. [PMID: 37985436 PMCID: PMC10805491 DOI: 10.1111/jcmm.18028] [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: 05/02/2023] [Revised: 10/15/2023] [Accepted: 10/24/2023] [Indexed: 11/22/2023] Open
Abstract
Pathological cardiac hypertrophy is a key contributor to heart failure, and the molecular mechanisms underlying honokiol (HNK)-mediated cardioprotection against this condition remain worth further exploring. This study aims to investigate the effect of HNK on angiotensin II (Ang II)-induced myocardial hypertrophy and elucidate the underlying mechanisms. Sprague-Dawley rats were exposed to Ang II infusion, followed by HNK or vehicle treatment for 4 weeks. Our results showed that HNK treatment protected against Ang II-induced myocardial hypertrophy, fibrosis and dysfunction in vivo and inhibited Ang II-induced hypertrophy in neonatal rat ventricular myocytes in vitro. Mechanistically, HNK suppressed the Ang II-induced Nur77 expression at the transcriptional level and promoted ubiquitination-mediated degradation of Nur77, leading to dissociation of the Nur77-LKB1 complex. This facilitated the translocation of LKB1 into the cytoplasm and activated the LKB1-AMPK pathway. Our findings suggest that HNK attenuates pathological remodelling and cardiac dysfunction induced by Ang II by promoting dissociation of the Nur77-LKB1 complex and subsequent activation of AMPK signalling. This study uncovers a novel role of HNK on the LKB1-AMPK pathway to protect against cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaoyan Lin
- Echocardiological Department, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Hailin Zhang
- Cardiovascular Department, Fujian Institute of Hypertension, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yong Chu
- Cardiovascular Department, Fujian Institute of Hypertension, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yuze Zhang
- Cardiovascular Department, Fujian Institute of Hypertension, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Changsheng Xu
- Cardiovascular Department, Fujian Institute of Hypertension, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Hong Xie
- Cardiovascular Department, Fujian Institute of Hypertension, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Qinyun Ruan
- Echocardiological Department, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Jinxiu Lin
- Cardiovascular Department, Fujian Institute of Hypertension, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Chun‐Kai Huang
- Cardiovascular Department, Fujian Institute of Hypertension, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Dajun Chai
- Cardiovascular Department, Fujian Institute of Hypertension, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| |
Collapse
|
9
|
Yilmaz A, Liraz-Zaltsman S, Shohami E, Gordevičius J, Kerševičiūtė I, Sherman E, Bahado-Singh RO, Graham SF. The longitudinal biochemical profiling of TBI in a drop weight model of TBI. Sci Rep 2023; 13:22260. [PMID: 38097614 PMCID: PMC10721861 DOI: 10.1038/s41598-023-48539-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and disability worldwide, particularly among individuals under the age of 45. It is a complex, and heterogeneous disease with a multifaceted pathophysiology that remains to be elucidated. Metabolomics has the potential to identify metabolic pathways and unique biochemical profiles associated with TBI. Herein, we employed a longitudinal metabolomics approach to study TBI in a weight drop mouse model to reveal metabolic changes associated with TBI pathogenesis, severity, and secondary injury. Using proton nuclear magnetic resonance (1H NMR) spectroscopy, we biochemically profiled post-mortem brain from mice that suffered mild TBI (N = 25; 13 male and 12 female), severe TBI (N = 24; 11 male and 13 female) and sham controls (N = 16; 11 male and 5 female) at baseline, day 1 and day 7 following the injury. 1H NMR-based metabolomics, in combination with bioinformatic analyses, highlights a few significant metabolites associated with TBI severity and perturbed metabolism related to the injury. We report that the concentrations of taurine, creatinine, adenine, dimethylamine, histidine, N-Acetyl aspartate, and glucose 1-phosphate are all associated with TBI severity. Longitudinal metabolic observation of brain tissue revealed that mild TBI and severe TBI lead distinct metabolic profile changes. A multi-class model was able to classify the severity of injury as well as time after TBI with estimated 86% accuracy. Further, we identified a high degree of correlation between respective hemisphere metabolic profiles (r > 0.84, p < 0.05, Pearson correlation). This study highlights the metabolic changes associated with underlying TBI severity and secondary injury. While comprehensive, future studies should investigate whether: (a) the biochemical pathways highlighted here are recapitulated in the brain of TBI sufferers and (b) if the panel of biomarkers are also as effective in less invasively harvested biomatrices, for objective and rapid identification of TBI severity and prognosis.
Collapse
Affiliation(s)
- Ali Yilmaz
- Metabolomics Department, Beaumont Research Institute, Beaumont Health, Royal Oak, MI, 48073, USA
- Oakland University-William Beaumont School of Medicine, Rochester, MI, 48073, USA
| | - Sigal Liraz-Zaltsman
- Department of Pharmacology, The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat-Gan, Israel
- Department of Sports Therapy, Institute for Health and Medical Professions, Ono Academic College, Qiryat Ono, Israel
| | - Esther Shohami
- Department of Pharmacology, The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Juozas Gordevičius
- VUGENE LLC, 625 EKenmoor Avenue Southeast, Suite 301, PMB 96578, Grand Rapids, MI, 49546, USA
| | - Ieva Kerševičiūtė
- VUGENE LLC, 625 EKenmoor Avenue Southeast, Suite 301, PMB 96578, Grand Rapids, MI, 49546, USA
| | - Eric Sherman
- Wayne State University School of Medicine, Detroit, MI, 48202, USA
| | - Ray O Bahado-Singh
- Oakland University-William Beaumont School of Medicine, Rochester, MI, 48073, USA
| | - Stewart F Graham
- Metabolomics Department, Beaumont Research Institute, Beaumont Health, Royal Oak, MI, 48073, USA.
- Oakland University-William Beaumont School of Medicine, Rochester, MI, 48073, USA.
| |
Collapse
|
10
|
Beggs MR, Ashkin A, Larsen BMK, Garros D. Measuring Energy Requirements of Traumatic Brain Injury Patients in Pediatric Intensive Care With Indirect Calorimetry: A Comparison With Empiric Methods. Pediatr Crit Care Med 2023; 24:e468-e475. [PMID: 37125799 DOI: 10.1097/pcc.0000000000003266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
OBJECTIVES Energy requirements following moderate or severe pediatric traumatic brain injury (TBI) have not been fully elucidated. Indirect calorimetry (IC) is the gold standard for measuring resting energy expenditure (MREE) in PICU. However, technical complexity limits its use. We aimed to determine whether MREE differs from standard of care energy estimation and delivery in a cohort of pediatric patients following moderate to severe TBI during PICU admission. DESIGN Retrospective case series study. SETTING Single-center, 16-bed general PICU in Canada between May 2011 and January 2019. PATIENTS Children (0-18 yr) admitted to a PICU for moderate (Glasgow Coma Scale [GCS] 9-12) to severe TBI (GCS < 9) and had an IC study performed while mechanically ventilated. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS During the study period, 245 patients were admitted with the diagnosis of trauma with TBI. The study includes a convenience sample of 26 patients with severe ( n = 23) and moderate ( n = 3) TBI who underwent a total of 34 IC measurements. MREE varied considerably from 29% to 144% of predicted energy expenditure. Using Bland-Altman comparative analysis, neither Schofield nor World Health Organization predictive equations were in agreement with MREE. Only one measurement revealed that the patient was appropriately fed (energy provided in nutrition support was within 10% of MREE); 10 (38%) measurements revealed overfeeding and 15 (58%) underfeeding at the time of testing. CONCLUSIONS The present study adds to the small body of literature highlighting the limitations of predictive equations to evaluate energy requirements following moderate to severe pediatric TBI. IC, when feasible, should be used as the preferred method to orient PICU teams to feed such vulnerable patients.
Collapse
Affiliation(s)
- Megan R Beggs
- Nutrition Services, Alberta Health Services, Edmonton, AB, Canada
- Women and Children's Health Research Institute (WCHRI), University of Alberta, Edmonton, AB, Canada
| | - Allison Ashkin
- Nutrition Services, Alberta Health Services, Edmonton, AB, Canada
- Pediatric Intensive Care Unit, Stollery Children's Hospital, Edmonton, AB, Canada
| | - Bodil M K Larsen
- Nutrition Services, Alberta Health Services, Edmonton, AB, Canada
- Pediatric Intensive Care Unit, Stollery Children's Hospital, Edmonton, AB, Canada
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
| | - Daniel Garros
- Pediatric Intensive Care Unit, Stollery Children's Hospital, Edmonton, AB, Canada
- Division of Critical Care, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
11
|
Strogulski NR, Portela LV, Polster BM, Loane DJ. Fundamental Neurochemistry Review: Microglial immunometabolism in traumatic brain injury. J Neurochem 2023; 167:129-153. [PMID: 37759406 PMCID: PMC10655864 DOI: 10.1111/jnc.15959] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Traumatic brain injury (TBI) is a devastating neurological disorder caused by a physical impact to the brain that promotes diffuse damage and chronic neurodegeneration. Key mechanisms believed to support secondary brain injury include mitochondrial dysfunction and chronic neuroinflammation. Microglia and brain-infiltrating macrophages are responsible for neuroinflammatory cytokine and reactive oxygen species (ROS) production after TBI. Their production is associated with loss of homeostatic microglial functions such as immunosurveillance, phagocytosis, and immune resolution. Beyond providing energy support, mitochondrial metabolic pathways reprogram the pro- and anti-inflammatory machinery in immune cells, providing a critical immunometabolic axis capable of regulating immunologic response to noxious stimuli. In the brain, the capacity to adapt to different environmental stimuli derives, in part, from microglia's ability to recognize and respond to changes in extracellular and intracellular metabolite levels. This capacity is met by an equally plastic metabolism, capable of altering immune function. Microglial pro-inflammatory activation is associated with decreased mitochondrial respiration, whereas anti-inflammatory microglial polarization is supported by increased oxidative metabolism. These metabolic adaptations contribute to neuroimmune responses, placing mitochondria as a central regulator of post-traumatic neuroinflammation. Although it is established that profound neurometabolic changes occur following TBI, key questions related to metabolic shifts in microglia remain unresolved. These include (a) the nature of microglial mitochondrial dysfunction after TBI, (b) the hierarchical positions of different metabolic pathways such as glycolysis, pentose phosphate pathway, glutaminolysis, and lipid oxidation during secondary injury and recovery, and (c) how immunometabolism alters microglial phenotypes, culminating in chronic non-resolving neuroinflammation. In this basic neurochemistry review article, we describe the contributions of immunometabolism to TBI, detail primary evidence of mitochondrial dysfunction and metabolic impairments in microglia and macrophages, discuss how major metabolic pathways contribute to post-traumatic neuroinflammation, and set out future directions toward advancing immunometabolic phenotyping in TBI.
Collapse
Affiliation(s)
- Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Luis V. Portela
- Neurotrauma and Biomarkers Laboratory, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Brian M. Polster
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David J. Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Ma Y, Jiang Q, Yang B, Hu X, Shen G, Shen W, Xu J. Platelet mitochondria, a potent immune mediator in neurological diseases. Front Physiol 2023; 14:1210509. [PMID: 37719457 PMCID: PMC10502307 DOI: 10.3389/fphys.2023.1210509] [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: 04/24/2023] [Accepted: 08/17/2023] [Indexed: 09/19/2023] Open
Abstract
Dysfunction of the immune response is regarded as a prominent feature of neurological diseases, including neurodegenerative diseases, malignant tumors, acute neurotraumatic insult, and cerebral ischemic/hemorrhagic diseases. Platelets play a fundamental role in normal hemostasis and thrombosis. Beyond those normal functions, platelets are hyperactivated and contribute crucially to inflammation and immune responses in the central nervous system (CNS). Mitochondria are pivotal organelles in platelets and are responsible for generating most of the ATP that is used for platelet activation and aggregation (clumping). Notably, platelet mitochondria show marked morphological and functional alterations under heightened inflammatory/oxidative stimulation. Mitochondrial dysfunction not only leads to platelet damage and apoptosis but also further aggravates immune responses. Improving mitochondrial function is hopefully an effective strategy for treating neurological diseases. In this review, the authors discuss the immunomodulatory roles of platelet-derived mitochondria (PLT-mitos) in neurological diseases and summarize the neuroprotective effects of platelet mitochondria transplantation.
Collapse
Affiliation(s)
- Yan Ma
- Transfusion Research Department, Wuhan Blood Center, Wuhan, Hubei, China
- Institute of Blood Transfusion of Hubei Province, Wuhan Blood Center, Wuhan, Hubei, China
- Wuhan National Laboratory for Optoelectronics and School of Physics, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Jiang
- Transfusion Research Department, Wuhan Blood Center, Wuhan, Hubei, China
- Institute of Blood Transfusion of Hubei Province, Wuhan Blood Center, Wuhan, Hubei, China
- Wuhan National Laboratory for Optoelectronics and School of Physics, Huazhong University of Science and Technology, Wuhan, China
| | - Bingxin Yang
- Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoyu Hu
- Transfusion Research Department, Wuhan Blood Center, Wuhan, Hubei, China
- Institute of Blood Transfusion of Hubei Province, Wuhan Blood Center, Wuhan, Hubei, China
- Wuhan National Laboratory for Optoelectronics and School of Physics, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Shen
- Transfusion Research Department, Wuhan Blood Center, Wuhan, Hubei, China
- Institute of Blood Transfusion of Hubei Province, Wuhan Blood Center, Wuhan, Hubei, China
| | - Wei Shen
- Wuhan Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Xu
- Wuhan Blood Center, Wuhan, Hubei, China
| |
Collapse
|
13
|
Agrawal RR, Larrea D, Xu Y, Shi L, Zirpoli H, Cummins LG, Emmanuele V, Song D, Yun TD, Macaluso FP, Min W, Kernie SG, Deckelbaum RJ, Area-Gomez E. Alzheimer's-Associated Upregulation of Mitochondria-Associated ER Membranes After Traumatic Brain Injury. Cell Mol Neurobiol 2023; 43:2219-2241. [PMID: 36571634 PMCID: PMC10287820 DOI: 10.1007/s10571-022-01299-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 10/04/2022] [Indexed: 12/27/2022]
Abstract
Traumatic brain injury (TBI) can lead to neurodegenerative diseases such as Alzheimer's disease (AD) through mechanisms that remain incompletely characterized. Similar to AD, TBI models present with cellular metabolic alterations and modulated cleavage of amyloid precursor protein (APP). Specifically, AD and TBI tissues display increases in amyloid-β as well as its precursor, the APP C-terminal fragment of 99 a.a. (C99). Our recent data in cell models of AD indicate that C99, due to its affinity for cholesterol, induces the formation of transient lipid raft domains in the ER known as mitochondria-associated endoplasmic reticulum (ER) membranes ("MAM" domains). The formation of these domains recruits and activates specific lipid metabolic enzymes that regulate cellular cholesterol trafficking and sphingolipid turnover. Increased C99 levels in AD cell models promote MAM formation and significantly modulate cellular lipid homeostasis. Here, these phenotypes were recapitulated in the controlled cortical impact (CCI) model of TBI in adult mice. Specifically, the injured cortex and hippocampus displayed significant increases in C99 and MAM activity, as measured by phospholipid synthesis, sphingomyelinase activity and cholesterol turnover. In addition, our cell type-specific lipidomics analyses revealed significant changes in microglial lipid composition that are consistent with the observed alterations in MAM-resident enzymes. Altogether, we propose that alterations in the regulation of MAM and relevant lipid metabolic pathways could contribute to the epidemiological connection between TBI and AD.
Collapse
Affiliation(s)
- Rishi R Agrawal
- Institute of Human Nutrition, Columbia University Irving Medical Center, 630 W. 168th St., Presbyterian Hospital 15E-1512, New York, NY, 10032, USA.
- Denali Therapeutics Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| | - Delfina Larrea
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA
| | - Yimeng Xu
- Biomarkers Core Laboratory, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 W. 168th St., Presbyterian Hospital 10-105, New York, NY, 10032, USA
| | - Lingyan Shi
- Department of Chemistry, Columbia University, 3000 Broadway, Havemeyer Hall, New York, NY, 10027, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Hylde Zirpoli
- Institute of Human Nutrition, Columbia University Irving Medical Center, 630 W. 168th St., Presbyterian Hospital 15E-1512, New York, NY, 10032, USA
| | - Leslie G Cummins
- Analytical Imaging Facility, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, 10461, USA
| | - Valentina Emmanuele
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA
| | - Donghui Song
- Department of Chemistry, Columbia University, 3000 Broadway, Havemeyer Hall, New York, NY, 10027, USA
| | - Taekyung D Yun
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA
| | - Frank P Macaluso
- Analytical Imaging Facility, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, 10461, USA
| | - Wei Min
- Biomarkers Core Laboratory, Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 622 W. 168th St., Presbyterian Hospital 10-105, New York, NY, 10032, USA
| | - Steven G Kernie
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA
- Department of Pediatrics, Columbia University Irving Medical Center, 622 W. 168th St., Presbyterian Hospital 17, New York, NY, 10032, USA
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Columbia University Irving Medical Center, 630 W. 168th St., Presbyterian Hospital 15E-1512, New York, NY, 10032, USA
- Department of Pediatrics, Columbia University Irving Medical Center, 622 W. 168th St., Presbyterian Hospital 17, New York, NY, 10032, USA
| | - Estela Area-Gomez
- Institute of Human Nutrition, Columbia University Irving Medical Center, 630 W. 168th St., Presbyterian Hospital 15E-1512, New York, NY, 10032, USA.
- Department of Neurology, Neurological Institute, Columbia University Irving Medical Center, 710 W. 168th St., New York, NY, 10032, USA.
- Centro de Investigaciones Biológicas Margarita Salas - CSIC, C. Ramiro de Maeztu, 9, 28040, Madrid, Spain.
| |
Collapse
|
14
|
Forti RM, Hobson LJ, Benson EJ, Ko TS, Ranieri NR, Laurent G, Weeks MK, Widmann NJ, Morton S, Davis AM, Sueishi T, Lin Y, Wulwick KS, Fagan N, Shin SS, Kao SH, Licht DJ, White BR, Kilbaugh TJ, Yodh AG, Baker WB. Non-invasive diffuse optical monitoring of cerebral physiology in an adult swine-model of impact traumatic brain injury. BIOMEDICAL OPTICS EXPRESS 2023; 14:2432-2448. [PMID: 37342705 PMCID: PMC10278631 DOI: 10.1364/boe.486363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/17/2023] [Accepted: 04/12/2023] [Indexed: 06/23/2023]
Abstract
In this study, we used diffuse optics to address the need for non-invasive, continuous monitoring of cerebral physiology following traumatic brain injury (TBI). We combined frequency-domain and broadband diffuse optical spectroscopy with diffuse correlation spectroscopy to monitor cerebral oxygen metabolism, cerebral blood volume, and cerebral water content in an established adult swine-model of impact TBI. Cerebral physiology was monitored before and after TBI (up to 14 days post injury). Overall, our results suggest that non-invasive optical monitoring can assess cerebral physiologic impairments post-TBI, including an initial reduction in oxygen metabolism, development of cerebral hemorrhage/hematoma, and brain swelling.
Collapse
Affiliation(s)
- Rodrigo M. Forti
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
| | - Lucas J. Hobson
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Emilie J. Benson
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tiffany S. Ko
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicolina R. Ranieri
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
| | - Gerard Laurent
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
| | - M. Katie Weeks
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicholas J. Widmann
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sarah Morton
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anthony M. Davis
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Takayuki Sueishi
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yuxi Lin
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Karli S. Wulwick
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicholas Fagan
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Samuel S. Shin
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shih-Han Kao
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Daniel J. Licht
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brian R. White
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Cardiology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Todd J. Kilbaugh
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arjun G. Yodh
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wesley B. Baker
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Resuscitation Science Center of Emphasis, CHOP Research Institute, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Hemispheric analysis of mitochondrial Complex I and II activity in the mouse model of ischemia-reperfusion-induced injury. Mitochondrion 2023; 69:147-158. [PMID: 36764500 DOI: 10.1016/j.mito.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023]
Abstract
Brain ischemia/reperfusion injury results in a variable mixture of cellular damage, but little is known about possible patterns of mitochondrial dysfunction from the scope of hemispheric processes. The current study used high-resolution fluorespirometry to compare ipsi- and contralateral hemispheres' linked respiration and ROS emission after 60-minutes of filament induced middle cerebral artery occlusion (fMCAo) and 2, 24, 72, and 168 h after reperfusion in mice. Our findings highlight that experimental ischemic stroke resulted in higher mitochondrial respiration in the contralateral compared to the ipsilateral hemisphere and highest ROS emission in ipsilateral hemisphere. The largest difference between the ipsilateral and contralateral hemispheres was observed 2 h after reperfusion in Complex I and II ETS state. Oxygen flux returns to near baseline 72 h after reperfusion without any changes thereafter in Complex I and II respiration. Studying the effects of brain mitochondrial functionality after ischemic stroke in each cerebral hemisphere separately provides a better understanding about the molecular and compensatory processes of the contralateral hemisphere, a region of the brain often neglected in stroke research.
Collapse
|
16
|
Shin SS, Chawla S, Jang DH, Mazandi VM, Weeks MK, Kilbaugh TJ. Imaging of White Matter Injury Correlates with Plasma and Tissue Biomarkers in Pediatric Porcine Model of Traumatic Brain Injury. J Neurotrauma 2023; 40:74-85. [PMID: 35876453 PMCID: PMC9917326 DOI: 10.1089/neu.2022.0178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Traumatic brain injury (TBI) causes significant white matter injury, which has been characterized by various rodent and human clinical studies. The exact time course of imaging changes in a pediatric brain after TBI and its relation to biomarkers of injury and cellular function, however, is unknown. To study the changes in major white matter structures using a valid model of TBI that is comparable to a human pediatric brain in terms of size and anatomical features, we utilized a four-week-old pediatric porcine model of injury with controlled cortical impact (CCI). Using diffusion tensor imaging differential tractography, we show progressive anisotropy changes at major white matter tracts such as the corona radiata and inferior fronto-occipital fasciculus between day 1 and day 30 after injury. Moreover, correlational tractography shows a large part of bilateral corona radiata having positive correlation with the markers of cellular respiration. In contrast, bilateral corona radiata has a negative correlation with the plasma biomarkers of injury such as neurofilament light or glial fibrillary acidic protein. These are expected correlational findings given that higher integrity of white matter would be expected to correlate with lower injury biomarkers. We then studied the magnetic resonance spectroscopy findings and report decrease in a N-acetylaspartate/creatinine (NAA/Cr) ratio at the pericontusional cortex, subcortical white matter, corona radiata, thalamus, genu, and splenium of corpus callosum at 30 days indicating injury. There was also an increase in choline/creatinine ratio in these regions indicating rapid membrane turnover. Given the need for a pediatric TBI model that is comparable to human pediatric TBI, these data support the use of a pediatric pig model with CCI in future investigations of therapeutic agents. This model will allow future TBI researchers to rapidly translate our pre-clinical study findings into clinical trials for pediatric TBI.
Collapse
Affiliation(s)
- Samuel S. Shin
- Division of Neurocritical Care, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David H. Jang
- Department of Emergency Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vanessa M. Mazandi
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - M. Katie Weeks
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Todd J. Kilbaugh
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Hwang M, Chattaraj R, Sridharan A, Shin SS, Viaene AN, Haddad S, Khrichenko D, Sehgal C, Lee D, Kilbaugh TJ. Can Ultrasound-Guided Xenon Delivery Provide Neuroprotection in Traumatic Brain Injury? Neurotrauma Rep 2022; 3:97-104. [PMID: 35317306 PMCID: PMC8935480 DOI: 10.1089/neur.2021.0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with high mortality and morbidity in children and adults. Unfortunately, there is no effective management for TBI in the acute setting. Rodent studies have shown that xenon, a well-known anesthetic gas, can be neuroprotective when administered post-TBI. Gas inhalation therapy, however, the approach typically used for administering xenon, is expensive, inconvenient, and fraught with systemic side effects. Therapeutic delivery to the brain is minimal, with much of the inhaled gas cleared by the lungs. To bridge major gaps in clinical care and enhance cerebral delivery of xenon, this study introduces a novel xenon delivery technique, utilizing microbubbles, in which a high impulse ultrasound signal is used for targeted cerebral release of xenon. Briefly, an ultrasound pulse is applied along the carotid artery at the level of the neck on intravenous injection of xenon microbubbles (XeMBs) resulting in release of xenon from microbubbles into the brain. This delivery technique employs a hand-held, portable ultrasound system that could be adopted in resource-limited environments. Using a high-fidelity porcine model, this study demonstrates the neuroprotective efficacy of xenon microbubbles in TBI for the first time.
Collapse
Affiliation(s)
- Misun Hwang
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rajarshi Chattaraj
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anush Sridharan
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Samuel S. Shin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Angela N. Viaene
- Department of Pathology, and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sophie Haddad
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Dmitry Khrichenko
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Chandra Sehgal
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daeyeon Lee
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Todd J. Kilbaugh
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Sowers JL, Sowers ML, Shavkunov AS, Hawkins BE, Wu P, DeWitt DS, Prough DS, Zhang K. Traumatic brain injury induces region-specific glutamate metabolism changes as measured by multiple mass spectrometry methods. iScience 2021; 24:103108. [PMID: 34622161 PMCID: PMC8479783 DOI: 10.1016/j.isci.2021.103108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 06/14/2021] [Accepted: 09/08/2021] [Indexed: 11/02/2022] Open
Abstract
The release of excess glutamate following traumatic brain injury (TBI) results in glutamate excitotoxicity and metabolic energy failure. Endogenous mechanisms for reducing glutamate concentration in the brain parenchyma following TBI are poorly understood. Using multiple mass spectrometry approaches, we examined TBI-induced changes to glutamate metabolism. We present evidence that glutamate concentration can be reduced by glutamate oxidation via a "truncated" tricarboxylic acid cycle coupled to the urea cycle. This process reduces glutamate levels, generates carbon for energy metabolism, leads to citrulline accumulation, and produces nitric oxide. Several key metabolites are identified by metabolomics in support of this mechanism and the locations of these metabolites in the injured hemisphere are demonstrated by MALDI-MS imaging. The results of this study establish the advantages of multiple mass spectrometry approaches and provide insights into glutamate metabolism following TBI that could lead to improved treatment approaches.
Collapse
Affiliation(s)
- James L Sowers
- MD-PhD Combined Degree Program, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mark L Sowers
- MD-PhD Combined Degree Program, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alexander S Shavkunov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bridget E Hawkins
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA.,Research Innovation and Scientific Excellence (RISE) Center, School of Nursing, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ping Wu
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Douglas S DeWitt
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kangling Zhang
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.,The Moody Project for Translational Traumatic Brain Injury Research, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
19
|
Zhou Z, Zhang K, Liu Z, Gao X, Huang K, Chen C, Wang D, Yang Q, Long Q. ATPAF1 deficiency impairs ATP synthase assembly and mitochondrial respiration. Mitochondrion 2021; 60:129-141. [PMID: 34375736 DOI: 10.1016/j.mito.2021.08.005] [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] [Received: 03/03/2021] [Revised: 07/08/2021] [Accepted: 08/05/2021] [Indexed: 11/28/2022]
Abstract
ATP11p and ATP12p are two nuclear-encoded mitochondrial chaperone proteins required for assembling the F1Fo-ATP synthase F1 sector. ATPAF1 and ATPAF2 are the mammalian homologs of ATP11p and ATP12p. However, the biochemical and physiological relevance of ATPAF1 and ATPAF2 in animal tissues with high energy-dependence remains unclear. To explore the in vivo role of ATP assembly and the effects of ATP synthase deficiency in animals, we have generated knockout (KO) mouse models of these assembly factors using CRISPR/Cas9 technology. While the Atpaf2-KO mice were embryonically lethal, Atpaf1-KO mice grew to adulthood but with smaller body sizes and elevated blood lactate later in life. We specifically investigated how ATPAF1 deficiency may affect ATP synthase biogenesis and mitochondrial respiration in the mouse heart, an organ highly energy-dependent. Western blots and Blue-Native electrophoresis (BN-PAGE) demonstrated a decreased F1 content and ATP synthase dimers in the Atpaf1-KO heart. Mitochondria from ATPAF1-deficient hearts showed ultrastructural abnormalities with condensed degenerated mitochondria, loss of cristae, and impaired respiratory capacity. ATP synthase deficiency also leads to impaired autophagy and mitochondrial dynamic. Consequently, decreased cardiac function was exhibited in adult Atpaf1-KO mice. The results provide strong support that ATPAF1 is essential for ATP synthase assembly and mitochondrial oxidative phosphorylation, thus playing a crucial role in maintaining cardiac structure and function in animals.
Collapse
Affiliation(s)
- Zhou Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China
| | - Kailiang Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China
| | - Zhiheng Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China
| | - Xu Gao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China
| | - Kai Huang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China
| | - Qinglin Yang
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Science Center-New Orleans, LA 70112, United States; Department of Nutrition Science, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| | - Qinqiang Long
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, China; Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Nutrition Science, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
20
|
Volk LE, Mavroudis CD, Ko T, Hallowell T, Delso N, Roberts AL, Starr J, Landis W, Lin Y, Hefti M, Morgan RW, Melchior RW, Rosenthal TM, Chappell A, Fisher D, Dreher M, Licht DJ, Chen J, Gaynor JW, Mascio CE, Kilbaugh TJ. Increased cerebral mitochondrial dysfunction and reactive oxygen species with cardiopulmonary bypass. Eur J Cardiothorac Surg 2021; 59:1256-1264. [PMID: 33367535 DOI: 10.1093/ejcts/ezaa439] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/26/2020] [Accepted: 11/04/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Neurodevelopmental injury after cardiac surgery using cardiopulmonary bypass (CPB) for congenital heart defects is common, but the mechanism behind this injury is unclear. This study examines the impact of CPB on cerebral mitochondrial reactive oxygen species (ROS) generation and mitochondrial bioenergetics. METHODS Twenty-three piglets (mean weight 4.2 ± 0.5 kg) were placed on CPB for either 1, 2, 3 or 4 h (n = 5 per group) or underwent anaesthesia without CPB (sham, n = 3). Microdialysis was used to measure metabolic markers of ischaemia. At the conclusion of CPB or 4 h of sham, brain tissue was harvested. Utilizing high-resolution respirometry, with simultaneous fluorometric analysis, mitochondrial respiration and ROS were measured. RESULTS There were no significant differences in markers of ischaemia between sham and experimental groups. Sham animals had significantly higher mitochondrial respiration than experimental animals, including maximal oxidative phosphorylation capacity of complex I (OXPHOSCI) (3.25 ± 0.18 vs 4-h CPB: 1.68 ± 0.10, P < 0.001) and maximal phosphorylating respiration capacity via convergent input through complexes I and II (OXPHOSCI+CII) (7.40 ± 0.24 vs 4-h CPB: 3.91 ± 0.20, P < 0.0001). At 4-h, experimental animals had significantly higher ROS related to non-phosphorylating respiration through complexes I and II (ETSCI+CII) than shams (1.08 ± 0.13 vs 0.64 ± 0.04, P = 0.026). CONCLUSIONS Even in the absence of local markers of ischaemia, CPB is associated with decreased mitochondrial respiration relative to shams irrespective of duration. Exposure to 4 h of CPB resulted in a significant increase in cerebral mitochondrial ROS formation compared to shorter durations. Further study is needed to improve the understanding of cerebral mitochondrial health and its effects on the pathophysiology of neurological injury following exposure to CPB.
Collapse
Affiliation(s)
- Lindsay E Volk
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Constantine D Mavroudis
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tiffany Ko
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Thomas Hallowell
- Division of Anesthesia and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nile Delso
- Division of Anesthesia and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anna L Roberts
- Division of Anesthesia and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan Starr
- Division of Anesthesia and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - William Landis
- Division of Anesthesia and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuxi Lin
- Division of Anesthesia and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marco Hefti
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Ryan W Morgan
- Division of Anesthesia and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Richard W Melchior
- Division of Perfusion Services, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tami M Rosenthal
- Division of Perfusion Services, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alexander Chappell
- Division of Perfusion Services, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Douglas Fisher
- Division of Perfusion Services, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Molly Dreher
- Division of Perfusion Services, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Daniel J Licht
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan Chen
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - J William Gaynor
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christopher E Mascio
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Todd J Kilbaugh
- Division of Anesthesia and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
21
|
Yu J, Meng F, He F, Chen F, Bao W, Yu Y, Zhou J, Gao J, Li J, Yao Y, Ge WP, Luo B. Metabolic Abnormalities in Patients with Chronic Disorders of Consciousness. Aging Dis 2021; 12:386-403. [PMID: 33815872 PMCID: PMC7990357 DOI: 10.14336/ad.2020.0812] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
The vegetative state (VS) and minimally conscious state (MCS) are two major types of chronic disorders of consciousness (DoC). The assessment of these two consciousness states generally relies on the Coma Recovery Scale-Revised (CRS-R) score, but a high misdiagnosis rate limits the generalized use of this score. To identify metabolites in human plasma that can accurately distinguish VS from MCS patients, comprehensive plasma metabolic profiles were obtained with targeted metabolomics analysis and untargeted and targeted lipidomics analysis. Univariate and multivariate analyses were used to assess the significance of differences. Compared with healthy controls (HCs), the DoC groups, Emerged from Minimally Conscious State (EMCS) group and Alzheimer’s disease (AD) group had significantly different metabolic profiles. Purine metabolism pathway differed the most between the DoC (MCS and VS) and HC groups. In this pathway, adenosine, ADP, and AMP, which are the derived products of ATP degradation, were decreased in the MCS and VS groups compared to healthy controls. More importantly, we identified certain lipids for which the levels were enriched in the VS or MCS groups. Specifically, phosphatidylcholine, (38:5)-H (PC(38:5)-H), and arachidonic acid (AA) differed substantially between the VS and MCS groups and may be used to distinguish these two groups of patients. Together, our findings suggest that metabolic profiling is significantly altered in patients with chronic DoC.
Collapse
Affiliation(s)
- Jie Yu
- 1Department of Neurology and Brain Medical Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Fanxia Meng
- 1Department of Neurology and Brain Medical Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Fangping He
- 1Department of Neurology and Brain Medical Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Fei Chen
- 2Children's Research Institute, Department of Neuroscience, University of Texas, Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wangxiao Bao
- 1Department of Neurology and Brain Medical Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yamei Yu
- 1Department of Neurology and Brain Medical Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jintao Zhou
- 1Department of Neurology and Brain Medical Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jian Gao
- 3Department of Rehabilitation, Hangzhou Hospital of Zhejiang Armed Police Corps, Hangzhou 310051, China
| | - Jingqi Li
- 3Department of Rehabilitation, Hangzhou Hospital of Zhejiang Armed Police Corps, Hangzhou 310051, China
| | - Yao Yao
- 4Department of Pharmaceutical and Biomedical Sciences, University of Georgia, GA 30602, USA
| | - Woo-Ping Ge
- 5Chinese Institute for Brain Research, Beijing 102206, China
| | - Benyan Luo
- 1Department of Neurology and Brain Medical Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
22
|
Morgan RW, Sutton RM, Himebauch AS, Roberts AL, Landis WP, Lin Y, Starr J, Ranganathan A, Delso N, Mavroudis CD, Volk L, Slovis J, Marquez AM, Nadkarni VM, Hefti M, Berg RA, Kilbaugh TJ. A randomized and blinded trial of inhaled nitric oxide in a piglet model of pediatric cardiopulmonary resuscitation. Resuscitation 2021; 162:274-283. [PMID: 33766668 DOI: 10.1016/j.resuscitation.2021.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 01/17/2023]
Abstract
AIM Inhaled nitric oxide (iNO) during cardiopulmonary resuscitation (CPR) improved systemic hemodynamics and outcomes in a preclinical model of adult in-hospital cardiac arrest (IHCA) and may also have a neuroprotective role following cardiac arrest. The primary objectives of this study were to determine if iNO during CPR would improve cerebral hemodynamics and mitochondrial function in a pediatric model of lipopolysaccharide-induced shock-associated IHCA. METHODS After lipopolysaccharide infusion and ventricular fibrillation induction, 20 1-month-old piglets received hemodynamic-directed CPR and were randomized to blinded treatment with or without iNO (80 ppm) during and after CPR. Defibrillation attempts began at 10 min with a 20-min maximum CPR duration. Cerebral tissue from animals surviving 1-h post-arrest underwent high-resolution respirometry to evaluate the mitochondrial electron transport system and immunohistochemical analyses to assess neuropathology. RESULTS During CPR, the iNO group had higher mean aortic pressure (41.6 ± 2.0 vs. 36.0 ± 1.4 mmHg; p = 0.005); diastolic BP (32.4 ± 2.4 vs. 27.1 ± 1.7 mmHg; p = 0.03); cerebral perfusion pressure (25.0 ± 2.6 vs. 19.1 ± 1.8 mmHg; p = 0.02); and cerebral blood flow relative to baseline (rCBF: 243.2 ± 54.1 vs. 115.5 ± 37.2%; p = 0.02). Among the 8/10 survivors in each group, the iNO group had higher mitochondrial Complex I oxidative phosphorylation in the cerebral cortex (3.60 [3.56, 3.99] vs. 3.23 [2.44, 3.46] pmol O2/s mg; p = 0.01) and hippocampus (4.79 [4.35, 5.18] vs. 3.17 [2.75, 4.58] pmol O2/s mg; p = 0.02). There were no other differences in mitochondrial respiration or brain injury between groups. CONCLUSIONS Treatment with iNO during CPR resulted in superior systemic hemodynamics, rCBF, and cerebral mitochondrial Complex I respiration in this pediatric cardiac arrest model.
Collapse
Affiliation(s)
- Ryan W Morgan
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States; Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, United States.
| | - Robert M Sutton
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States; Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, United States
| | - Adam S Himebauch
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States; Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, United States
| | - Anna L Roberts
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States
| | - William P Landis
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States
| | - Yuxi Lin
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States
| | - Jonathan Starr
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States
| | - Abhay Ranganathan
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States
| | - Nile Delso
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States
| | - Constantine D Mavroudis
- Department of Surgery, Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, United States
| | - Lindsay Volk
- Department of Surgery, Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, United States
| | - Julia Slovis
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States
| | - Alexandra M Marquez
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States
| | - Vinay M Nadkarni
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States; Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, United States
| | - Marco Hefti
- Department of Pathology, University of Iowa Carver College of Medicine, United States
| | - Robert A Berg
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States; Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, United States
| | - Todd J Kilbaugh
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, United States; Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine at the University of Pennsylvania, United States
| |
Collapse
|
23
|
Datzmann T, Kapapa T, Scheuerle A, McCook O, Merz T, Unmuth S, Hoffmann A, Mathieu R, Mayer S, Mauer UM, Röhrer S, Yilmazer-Hanke D, Möller P, Nussbaum BL, Calzia E, Gröger M, Hartmann C, Radermacher P, Wepler M. In-depth characterization of a long-term, resuscitated model of acute subdural hematoma-induced brain injury. J Neurosurg 2021; 134:223-234. [PMID: 31860806 DOI: 10.3171/2019.9.jns191789] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/13/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Acute subdural hematoma (ASDH) is a leading entity in brain injury. Rodent models mostly lack standard intensive care, while large animal models frequently are only short term. Therefore, the authors developed a long-term, resuscitated porcine model of ASDH-induced brain injury and report their findings. METHODS Anesthetized, mechanically ventilated, and instrumented pigs with human-like coagulation underwent subdural injection of 20 mL of autologous blood and subsequent observation for 54 hours. Continuous bilateral multimodal brain monitoring (intracranial pressure [ICP], cerebral perfusion pressure [CPP], partial pressure of oxygen in brain tissue [PbtO2], and brain temperature) was combined with intermittent neurological assessment (veterinary modified Glasgow Coma Scale [MGCS]), microdialysis, and measurement of plasma protein S100β, GFAP, neuron-specific enolase [NSE], nitrite+nitrate, and isoprostanes. Fluid resuscitation and continuous intravenous norepinephrine were targeted to maintain CPP at pre-ASDH levels. Immediately postmortem, the brains were taken for macroscopic and histological evaluation, immunohistochemical analysis for nitrotyrosine formation, albumin extravasation, NADPH oxidase 2 (NOX2) and GFAP expression, and quantification of tissue mitochondrial respiration. RESULTS Nine of 11 pigs survived the complete observation period. While ICP significantly increased after ASDH induction, CPP, PbtO2, and the MGCS score remained unaffected. Blood S100β levels significantly fell over time, whereas GFAP, NSE, nitrite+nitrate, and isoprostane concentrations were unaltered. Immunohistochemistry showed nitrotyrosine formation, albumin extravasation, NOX2 expression, fibrillary astrogliosis, and microglial activation. CONCLUSIONS The authors describe a clinically relevant, long-term, resuscitated porcine model of ASDH-induced brain injury. Despite the morphological injury, maintaining CPP and PbtO2 prevented serious neurological dysfunction. This model is suitable for studying therapeutic interventions during hemorrhage-induced acute brain injury with standard brain-targeted intensive care.
Collapse
Affiliation(s)
- Thomas Datzmann
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
- 2Klinik für Anästhesiologie
| | | | | | - Oscar McCook
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Tamara Merz
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Sarah Unmuth
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Andrea Hoffmann
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - René Mathieu
- 5Klinik für Neurochirurgie, Bundeswehrkrankenhaus Ulm; and
| | - Simon Mayer
- 5Klinik für Neurochirurgie, Bundeswehrkrankenhaus Ulm; and
| | - Uwe Max Mauer
- 5Klinik für Neurochirurgie, Bundeswehrkrankenhaus Ulm; and
| | - Stefan Röhrer
- 6Abteilung für Neurochirurgie, Klinikum Aalen, Germany
| | | | - Peter Möller
- 8Institut für Pathologie, Universitätsklinikum, Ulm
| | - Benedikt Lukas Nussbaum
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
- 2Klinik für Anästhesiologie
| | - Enrico Calzia
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Michael Gröger
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Clair Hartmann
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
- 2Klinik für Anästhesiologie
| | - Peter Radermacher
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
| | - Martin Wepler
- 1Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung
- 2Klinik für Anästhesiologie
| |
Collapse
|
24
|
Kang W, Yamatoya K, Miyado K, Miyado M, Miyamoto Y. Neuronal expression of Ca 2+ oscillation initiator is linked to rapid neonatal growth in mice. MICROPUBLICATION BIOLOGY 2020; 2020. [PMID: 33274323 PMCID: PMC7704253 DOI: 10.17912/micropub.biology.000325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Woojin Kang
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan
| | - Kenji Yamatoya
- Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Chiba 279-0021, Japan
| | - Kenji Miyado
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan
| | - Mami Miyado
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan
| | - Yoshitaka Miyamoto
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya, Tokyo 157-8535, Japan
| |
Collapse
|
25
|
Underwood E, Redell JB, Zhao J, Moore AN, Dash PK. A method for assessing tissue respiration in anatomically defined brain regions. Sci Rep 2020; 10:13179. [PMID: 32764697 PMCID: PMC7413397 DOI: 10.1038/s41598-020-69867-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/16/2020] [Indexed: 12/28/2022] Open
Abstract
The survival and function of brain cells requires uninterrupted ATP synthesis. Different brain structures subserve distinct neurological functions, and therefore have different energy production/consumption requirements. Typically, mitochondrial function is assessed following their isolation from relatively large amounts of starting tissue, making it difficult to ascertain energy production/failure in small anatomical locations. In order to overcome this limitation, we have developed and optimized a method to measure mitochondrial function in brain tissue biopsy punches excised from anatomically defined brain structures, including white matter tracts. We describe the procedures for maintaining tissue viability prior to performing the biopsy punches, as well as provide guidance for optimizing punch size and the drug doses needed to assess various aspects of mitochondrial respiration. We demonstrate that our method can be used to measure mitochondrial respiration in anatomically defined subfields within the rat hippocampus. Using this method, we present experimental results which show that a mild traumatic brain injury (mTBI, often referred to as concussion) causes differential mitochondrial responses within these hippocampal subfields and the corpus callosum, novel findings that would have been difficult to obtain using traditional mitochondrial isolation methods. Our method is easy to implement and will be of interest to researchers working in the field of brain bioenergetics and brain diseases.
Collapse
Affiliation(s)
- Erica Underwood
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - John B Redell
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA.
| |
Collapse
|
26
|
Guanosine protects against behavioural and mitochondrial bioenergetic alterations after mild traumatic brain injury. Brain Res Bull 2020; 163:31-39. [PMID: 32681970 DOI: 10.1016/j.brainresbull.2020.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) constitutes a heterogeneous cerebral insult induced by traumatic biomechanical forces. Mitochondria play a critical role in brain bioenergetics, and TBI induces several consequences related with oxidative stress and excitotoxicity clearly demonstrated in different experimental model involving TBI. Mitochondrial bioenergetics alterations can present several targets for therapeutics which could help reduce secondary brain lesions such as neuropsychiatric problems, including memory loss and motor impairment. Guanosine (GUO), an endogenous neuroprotective nucleoside, affords the long-term benefits of controlling brain neurodegeneration, mainly due to its capacity to activate the antioxidant defense system and maintenance of the redox system. However, little is known about the exact protective mechanism exerted by GUO on mitochondrial bioenergetics disruption induced by TBI. Thus, the aim of this study was to investigate the effects of GUO in brain cortical and hippocampal mitochondrial bioenergetics in the mild TBI model. Additionally, we aimed to assess whether mitochondrial damage induced by TBI may be related to behavioral alterations in rats. Our findings showed that 24 h post-TBI, GUO treatment promotes an adaptive response of mitochondrial respiratory chain increasing oxygen flux which it was able to protect against the uncoupling of oxidative phosphorylation (OXPHOS) induced by TBI, restored the respiratory electron transfer system (ETS) established with an uncoupler. Guanosine treatment also increased respiratory control ratio (RCR), an indicator of the state of mitochondrial coupling, which is related to the mitochondrial functionality. In addition, mitochondrial bioenergetics failure was closely related with locomotor, exploratory and memory impairments. The present study suggests GUO treatment post mild TBI could increase GDP endogenous levels and consequently increasing ATP levels promotes an increase of RCR increasing OXPHOS and in substantial improve mitochondrial respiration in different brain regions, which, in turn, could promote an improvement in behavioral parameters associated to the mild TBI. These findings may contribute to the development of future therapies with a target on failure energetic metabolism induced by TBI.
Collapse
|
27
|
Severo L, Godinho D, Machado F, Hartmann D, Fighera MR, Soares FA, Furian AF, Oliveira MS, Royes LF. The role of mitochondrial bioenergetics and oxidative stress in depressive behavior in recurrent concussion model in mice. Life Sci 2020; 257:117991. [PMID: 32569782 DOI: 10.1016/j.lfs.2020.117991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a public health problem in which even though 80 to 90% of cases are considered mild, usually starts a sequence of neurological disorders that can last a considerable time. Most of the research of this injury has been focused on oxidative stress and functional deficits; however, mechanisms that underlie the development of neuropsychiatric disorders remain little researched. Due to this, the present authors decided to investigate whether recurrent concussion protocols alter depressive-like phenotype behavior, and whether mitochondria play an indispensable role in this behavior or not. The experimental data revealed, for the first time, that the present protocol of recurrent concussions (4, 7, and 10 injuries) in mice did not alter immobility time during tail suspension tests (TSTs), but decreased hippocampal mitochondrial respiration and increased expression of proteins such as nuclear factor erythroid 2-related factor 2 (Nrf2) and superoxide (SOD2). This experimental data suggests that bioenergetic changes elicited by recurrent concussion did not induce depressive-like behavior, but activated the transcription factor of responsive antioxidant elements (ARE) that delay or prevent secondary cascades in this neurological disease.
Collapse
Affiliation(s)
- Leandro Severo
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica Universidade Federal de Santa Maria, 97105-900, Brazil; Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil.
| | - Douglas Godinho
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica Universidade Federal de Santa Maria, 97105-900, Brazil; Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Felipe Machado
- Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Diane Hartmann
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, 97105-900, Brazil
| | - Michele Rechia Fighera
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica Universidade Federal de Santa Maria, 97105-900, Brazil; Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Félix Alexandre Soares
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, 97105-900, Brazil
| | - Ana Flavia Furian
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Mauro Schneider Oliveira
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Luiz Fernando Royes
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica Universidade Federal de Santa Maria, 97105-900, Brazil; Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil.
| |
Collapse
|
28
|
Formoterol, a β 2-adrenoreceptor agonist, induces mitochondrial biogenesis and promotes cognitive recovery after traumatic brain injury. Neurobiol Dis 2020; 140:104866. [PMID: 32289370 DOI: 10.1016/j.nbd.2020.104866] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/12/2020] [Accepted: 04/05/2020] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) leads to acute necrosis at the site of injury followed by a sequence of secondary events lasting from hours to weeks and often years. Targeting mitochondrial impairment following TBI has shown improvements in brain mitochondrial bioenergetics and neuronal function. Recently formoterol, a highly selective β2-adrenoreceptor agonist, was found to induce mitochondrial biogenesis (MB) via Gβγ-Akt-eNOS-sGC pathway. Activation of MB is a novel approach that has been shown to restore mitochondrial function in several disease and injury models. We hypothesized that activation of MB as a target of formoterol after TBI would mitigate mitochondrial dysfunction, enhance neuronal function and improve behavioral outcomes. TBI-injured C57BL/6 male mice were injected (i.p.) with vehicle (normal saline) or formoterol (0.3 mg/kg) at 15 min, 8 h, 16 h, 24 h and then daily after controlled cortical impact (CCI) until euthanasia. After CCI, mitochondrial copy number and bioenergetic function were decreased in the ipsilateral cortex of the CCI-vehicle group. Compared to CCI-vehicle, cortical and hippocampal mitochondrial respiration rates as well as cortical mitochondrial DNA copy number were increased in the CCI-formoterol group. Mitochondrial Ca2+ buffering capacity in the hippocampus was higher in the CCI-formoterol group compared to CCI-vehicle group. Both assessments of cognitive performance, novel object recognition (NOR) and Morris water maze (MWM), decreased following CCI and were restored in the CCI-formoterol group. Although no changes were seen in the amount of cortical tissue spared between CCI-formoterol and CCI-vehicle groups, elevated levels of hippocampal neurons and improved white matter sparing in the corpus callosum were observed in CCI-formoterol group. Collectively, these results indicate that formoterol-mediated MB activation may be a potential therapeutic target to restore mitochondrial bioenergetics and promote functional recovery after TBI.
Collapse
|
29
|
Greco T, Vespa PM, Prins ML. Alternative substrate metabolism depends on cerebral metabolic state following traumatic brain injury. Exp Neurol 2020; 329:113289. [PMID: 32247790 PMCID: PMC8168752 DOI: 10.1016/j.expneurol.2020.113289] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 12/13/2022]
Abstract
Decreases in energy metabolism following traumatic brain injury (TBI) are attributed to impairment of glycolytic flux and oxidative phosphorylation. Glucose utilization post-TBI is decreased while administration of alternative substrates has been shown to be neuroprotective. Changes in energy metabolism following TBI happens in two phases; a period of hyper-metabolism followed by prolonged hypo-metabolism. It is not understood how different cerebral metabolic states may impact substrate metabolism and ultimately mitochondrial function. Adult male or female Sprague Dawley rats were given sham surgery or controlled cortical impact (CCI) and were assigned one of two administration schemes. Glucose, lactate or beta-hydroxybutyrate (BHB) were infused i.v. either starting immediately after injury or beginning 6 h post-injury for 3 h to reflect the hyper- and hypo-metabolic stages. Animals were euthanized 24 h post-injury. The peri-contusional cortex was collected and assayed for mitochondrial respiration peroxide production, and citrate synthase activity. Tissue acetyl-CoA, ATP, glycogen and HMGB1 were also quantified. Sex differences were observed in injury pattern. Administration based on cerebral metabolic state identified that only early lactate and late BHB improved mitochondrial function and peroxide production and TCA cycle intermediates in males. In contrast, both early and late BHB had deleterious effects on all aspects of metabolic measurements in females. These data stress there is no one optimal alternative substrate, but rather the fuel type used should be guided by both cerebral metabolic state and sex.
Collapse
Affiliation(s)
- Tiffany Greco
- UCLA Department of Neurosurgery, USA; UCLA Brain Injury Research Center, USA.
| | - Paul M Vespa
- UCLA Department of Neurosurgery, USA; UCLA Department of Neurology, USA
| | - Mayumi L Prins
- UCLA Department of Neurosurgery, USA; UCLA Interdepartmental Program for Neuroscience, USA; UCLA Brain Injury Research Center, USA
| |
Collapse
|
30
|
Abstract
Despite thousands of neuroprotectants demonstrating promise in preclinical trials, a neuroprotective therapeutic has yet to be approved for the treatment of acute brain injuries such as stroke or traumatic brain injury. Developing a more detailed understanding of models and populations demonstrating "neurological resilience" in spite of brain injury can give us important insights into new translational therapies. Resilience is the process of active adaptation to a stressor. In the context of neuroprotection, models of preconditioning and unique animal models of extreme physiology (such as hibernating species) reliably demonstrate resilience in the laboratory setting. In the clinical setting, resilience is observed in young patients and can be found in those with specific genetic polymorphisms. These important examples of resilience can help transform and extend the current neuroprotective framework from simply countering the injurious cascade into one that anticipates, monitors, and optimizes patients' physiological responses from the time of injury throughout the process of recovery. This review summarizes the underpinnings of key adaptations common to models of resilience and how this understanding can be applied to new neuroprotective approaches.
Collapse
Affiliation(s)
- Neel S Singhal
- Department of Neurology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA.
| | - Chung-Huan Sun
- Department of Neurology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| | - Evan M Lee
- Cardiovascular Research Institute, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
- Department of Physiology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| | - Dengke K Ma
- Cardiovascular Research Institute, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
- Department of Physiology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| |
Collapse
|
31
|
Using Serum Amino Acids to Predict Traumatic Brain Injury: A Systematic Approach to Utilize Multiple Biomarkers. Int J Mol Sci 2020; 21:ijms21051786. [PMID: 32150890 PMCID: PMC7084695 DOI: 10.3390/ijms21051786] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/17/2020] [Accepted: 03/02/2020] [Indexed: 01/06/2023] Open
Abstract
Traumatic brain injury (TBI) can cause biochemical and metabolomic alterations in the brain tissue and serum. These alterations can be used for diagnosis and prognosis of TBI. Here, the serum concentrations of seventeen amino acids (AA) were studied for their potential utility as biomarkers of TBI. Twenty-five female, 4-week-old piglets received diffuse (n = 13) or focal (n = 12) TBI. Blood samples were obtained both pre-injury and at either 24-h or 4-days post-TBI. To find a robust panel of biomarkers, the results of focal and diffuse TBIs were combined and multivariate logistic regression analysis, coupled with the best subset selection technique and repeated k-fold cross-validation method, was used to perform a thorough search of all possible subsets of AAs. The combination of serum glycine, taurine, and ornithine was optimal for TBI diagnosis, with 80% sensitivity and 86% overall prediction rate, and showed excellent TBI diagnostic performance, with 100% sensitivity and 78% overall prediction rate, on a separate validation dataset including four uninjured and five injured animals. We found that combinations of biomarkers outperformed any single biomarker. We propose this 3-AA serum biomarker panel to diagnose mild-to-moderate focal/diffuse TBI. The systematic approaches implemented herein can be used for combining parameters from various TBI assessments to develop/evaluate optimal multi-factorial diagnostic/prognostic TBI metrics.
Collapse
|
32
|
Ren YZ, Zhang BZ, Zhao XJ, Zhang ZY. Resolvin D1 ameliorates cognitive impairment following traumatic brain injury via protecting astrocytic mitochondria. J Neurochem 2020; 154:530-546. [PMID: 31951012 DOI: 10.1111/jnc.14962] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/29/2019] [Accepted: 01/02/2020] [Indexed: 12/18/2022]
Abstract
Cognitive impairment is one of the most common and devastating neuropsychiatric sequelae after traumatic brain injury (TBI), and hippocampal neuronal survival plays a causal role in this pathological process. Resolvin D1 (RvD1), an important endogenous specialized pro-resolving mediator, has recently been reported to exert a potent protective effect on mitochondria. This suggests that RvD1 may suppress neuroinflammation and protect astrocytic mitochondria at the same time to play further neuroprotective roles. C57BL/6 mice subjected to TBI using a controlled cortical impact device were used for in vivo experiments. Cultured primary mouse astrocytes and an N2a mouse neuroblastoma cell line were used for in vitro experiments. In TBI mice, RvD1 significantly ameliorated cognitive impairment, suppressed gliosis and alleviated neuronal loss in the hippocampus. To explore the mechanism underlying this activity, we verified that RvD1 can induce a higher level of mitophagy to remove damaged mitochondria and eliminate extra mitochondria-derived reactive oxygen species (mitoROS) by activating ALX4/FPR2 receptors in astrocytes. In an in vitro model, we further confirmed that RvD1 can protect mitochondrial morphology and membrane potential in astrocytes and thereby enhance the survival of neurons. Meanwhile, RvD1 was also shown to increase the expression of brain-derived neurotrophic factor and glutamate aspartate transporter in the hippocampus following TBI, which indicates a possible way by which RvD1 increases the supportive function of astrocytes. These findings suggest that RvD1 may be a potent therapeutic option for ameliorating cognitive impairment following TBI by controlling neuroinflammation and protecting astrocytic mitochondria.
Collapse
Affiliation(s)
- Yi-Zhi Ren
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Ben-Zheng Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xiao-Jing Zhao
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Zhi-Yuan Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, China.,The Key Laboratory of Antibody Technique of the Ministry of Health, Nanjing Medical University, Nanjing, China.,Department of Neurology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
33
|
Pyruvate Dehydrogenase and Tricarboxylic Acid Cycle Enzymes Are Sensitive Targets of Traumatic Brain Injury Induced Metabolic Derangement. Int J Mol Sci 2019; 20:ijms20225774. [PMID: 31744143 PMCID: PMC6888669 DOI: 10.3390/ijms20225774] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Using a closed-head impact acceleration model of mild or severe traumatic brain injury (mTBI or sTBI, respectively) in rats, we evaluated the effects of graded head impacts on the gene and protein expressions of pyruvate dehydrogenase (PDH), as well as major enzymes of mitochondrial tricarboxylic acid cycle (TCA). TBI was induced in anaesthetized rats by dropping 450 g from 1 (mTBI) or 2 m height (sTBI). After 6 h, 12 h, 24 h, 48 h, and 120 h gene expressions of enzymes and subunits of PDH. PDH kinases and phosphatases (PDK1-4 and PDP1-2, respectively), citrate synthase (CS), isocitrate dehydrogenase (IDH), oxoglutarate dehydrogenase (OGDH), succinate dehydrogenase (SDH), succinyl-CoA synthase (SUCLG), and malate dehydrogenase (MDH) were determined in whole brain extracts (n = 6 rats at each time for both TBI levels). In the same samples, the high performance liquid chromatographic (HPLC) determination of acetyl-coenzyme A (acetyl-CoA) and free coenzyme A (CoA-SH) was performed. Sham-operated animals (n = 6) were used as controls. After mTBI, the results indicated a general transient decrease, followed by significant increases, in PDH and TCA gene expressions. Conversely, permanent PDH and TCA downregulation occurred following sTBI. The inhibitory conditions of PDH (caused by PDP1-2 downregulations and PDK1-4 overexpression) and SDH appeared to operate only after sTBI. This produced almost no change in acetyl-CoA and free CoA-SH following mTBI and a remarkable depletion of both compounds after sTBI. These results again demonstrated temporary or steady mitochondrial malfunctioning, causing minimal or profound modifications to energy-related metabolites, following mTBI or sTBI, respectively. Additionally, PDH and SDH appeared to be highly sensitive to traumatic insults and are deeply involved in mitochondrial-related energy metabolism imbalance.
Collapse
|
34
|
Neuroprotective Effects of Methylene Blue In Vivo and In Vitro. Bull Exp Biol Med 2019; 167:455-459. [DOI: 10.1007/s10517-019-04548-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Indexed: 10/26/2022]
|
35
|
Pandya JD, Leung LY, Yang X, Flerlage WJ, Gilsdorf JS, Deng-Bryant Y, Shear DA. Comprehensive Profile of Acute Mitochondrial Dysfunction in a Preclinical Model of Severe Penetrating TBI. Front Neurol 2019; 10:605. [PMID: 31244764 PMCID: PMC6579873 DOI: 10.3389/fneur.2019.00605] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 05/22/2019] [Indexed: 01/11/2023] Open
Abstract
Mitochondria constitute a central role in brain energy metabolism, and play a pivotal role in the development of secondary pathophysiology and subsequent neuronal cell death following traumatic brain injury (TBI). Under normal circumstances, the brain consumes glucose as the preferred energy source for adenosine triphosphate (ATP) production over ketones. To understand the comprehensive picture of substrate-specific mitochondrial bioenergetics responses following TBI, adult male rats were subjected to either 10% unilateral penetrating ballistic-like brain injury (PBBI) or sham craniectomy (n = 5 animals per group). At 24 h post-injury, mitochondria were isolated from pooled brain regions (frontal cortex and striatum) of the ipsilateral hemisphere. Mitochondrial bioenergetics parameters were measured ex vivo in the presence of four sets of metabolic substrates: pyruvate+malate (PM), glutamate+malate (GM), succinate (Succ), and β-hydroxybutyrate+malate (BHBM). Additionally, mitochondrial matrix dehydrogenase activities [i.e., pyruvate dehydrogenase complex (PDHC), alpha-ketoglutarate dehydrogenase complex (α-KGDHC), and glutamate dehydrogenase (GDH)] and mitochondrial membrane-bound dehydrogenase activities [i.e., electron transport chain (ETC) Complex I, II, and IV] were compared between PBBI and sham groups. Furthermore, mitochondrial coenzyme contents, including NAD(t) and FAD(t), were quantitatively measured in both groups. Collectively, PBBI led to an overall significant decline in the ATP synthesis rates (43-50%; * p < 0.05 vs. sham) when measured using each of the four sets of substrates. The PDHC and GDH activities were significantly reduced in the PBBI group (42-53%; * p < 0.05 vs. sham), whereas no significant differences were noted in α-KGDHC activity between groups. Both Complex I and Complex IV activities were significantly reduced following PBBI (47-81%; * p < 0.05 vs. sham), whereas, Complex II activity was comparable between groups. The NAD(t) and FAD(t) contents were significantly decreased in the PBBI group (27-35%; * p < 0.05 vs. sham). The decreased ATP synthesis rates may be due to the significant reductions in brain mitochondrial dehydrogenase activities and coenzyme contents observed acutely following PBBI. These results provide a basis for the use of "alternative biofuels" for achieving higher ATP production following severe penetrating brain trauma.
Collapse
Affiliation(s)
- Jignesh D Pandya
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Lai Yee Leung
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Xiaofang Yang
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - William J Flerlage
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Janice S Gilsdorf
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Ying Deng-Bryant
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Deborah A Shear
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
36
|
Laouafa S, Roussel D, Marcouiller F, Soliz J, Gozal D, Bairam A, Joseph V. Roles of oestradiol receptor alpha and beta against hypertension and brain mitochondrial dysfunction under intermittent hypoxia in female rats. Acta Physiol (Oxf) 2019; 226:e13255. [PMID: 30635990 DOI: 10.1111/apha.13255] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/20/2022]
Abstract
AIM Chronic intermittent hypoxia (CIH) induces systemic (hypertension) and central alterations (mitochondrial dysfunction underlying cognitive deficits). We hypothesized that agonists of oestradiol receptors (ER) α and β prevent CIH-induced hypertension and brain mitochondrial dysfunction. METHODS Ovariectomized female rats were implanted with osmotic pumps delivering vehicle (Veh), the ERα agonist propylpyraoletriol (PPT - 30 μg/kg/day) or the ERβ agonist diarylpropionitril (DPN - 100 μg/kg/day). Animals were exposed to CIH (21%-10% FI O2 - 10 cycles/hour - 8 hours/day - 7 days) or normoxia. Arterial blood pressure was measured after CIH or normoxia exposures. Mitochondrial respiration and H2 O2 production were measured in brain cortex with high-resolution respirometry, as well as activity of complex I and IV of the electron transport chain, citrate synthase, pyruvate, and lactate dehydrogenase (PDH and LDH). RESULTS Propylpyraoletriol but not DPN prevented the rise of arterial pressure induced by CIH. CIH exposures decreased O2 consumption, complex I activity, and increased H2 O2 production. CIH had no effect on citrate synthase activity, but decreased PDH activity and increased LDH activity indicating higher anaerobic glycolysis. Propylpyraoletriol and DPN treatments prevented all these alterations. CONCLUSIONS We conclude that in OVX female rats, the ERα agonist prevents from CIH-induced hypertension while both ERα and ERβ agonists prevent the brain mitochondrial dysfunction and metabolic switch induced by CIH. These findings may have implications for menopausal women suffering of sleep apnoea regarding hormonal therapy.
Collapse
Affiliation(s)
- Sofien Laouafa
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| | - Damien Roussel
- CNRS, UMR 5023 Université Claude Bernard Lyon 1 Villeurbanne France
| | - François Marcouiller
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| | - Jorge Soliz
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| | - David Gozal
- Department of Child Health University of Missouri School of Medicine Columbia Missouri
| | - Aida Bairam
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| | - Vincent Joseph
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec Université Laval Quebec City Quebec Canada
| |
Collapse
|
37
|
Metabolic perturbations after pediatric TBI: It's not just about glucose. Exp Neurol 2019; 316:74-84. [PMID: 30951705 DOI: 10.1016/j.expneurol.2019.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/13/2019] [Accepted: 03/30/2019] [Indexed: 12/22/2022]
Abstract
Improved patient survival following pediatric traumatic brain injury (TBI) has uncovered a currently limited understanding of both the adaptive and maladaptive metabolic perturbations that occur during the acute and long-term phases of recovery. While much is known about the redundancy of metabolic pathways that provide adequate energy and substrates for normal brain growth and development, the field is only beginning to characterize perturbations in these metabolic pathways after pediatric TBI. To date, the majority of studies have focused on dysregulated oxidative glucose metabolism after injury; however, the immature brain is well-equipped to use alternative substrates to fuel energy production, growth, and development. A comprehensive understanding of metabolic changes associated with pediatric TBI cannot be limited to investigations of glucose metabolism alone. All energy substrates used by the brain should be considered in developing nutritional and pharmacological interventions for pediatric head trauma. This review summarizes post-injury changes in brain metabolism of glucose, lipids, ketone bodies, and amino acids with discussion of the therapeutic potential of altering substrate utilization to improve pediatric TBI outcomes.
Collapse
|
38
|
Chao H, Lin C, Zuo Q, Liu Y, Xiao M, Xu X, Li Z, Bao Z, Chen H, You Y, Kochanek PM, Yin H, Liu N, Kagan VE, Bayır H, Ji J. Cardiolipin-Dependent Mitophagy Guides Outcome after Traumatic Brain Injury. J Neurosci 2019; 39:1930-1943. [PMID: 30626699 PMCID: PMC6407296 DOI: 10.1523/jneurosci.3415-17.2018] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 11/21/2018] [Accepted: 12/28/2018] [Indexed: 01/14/2023] Open
Abstract
Mitochondrial energy production is essential for normal brain function. Traumatic brain injury (TBI) increases brain energy demands, results in the activation of mitochondrial respiration, associated with enhanced generation of reactive oxygen species. This chain of events triggers neuronal apoptosis via oxidation of a mitochondria-specific phospholipid, cardiolipin (CL). One pathway through which cells can avoid apoptosis is via elimination of damaged mitochondria by mitophagy. Previously, we showed that externalization of CL to the mitochondrial surface acts as an elimination signal in cells. Whether CL-mediated mitophagy occurs in vivo or its significance in the disease processes are not known. In this study, we showed that TBI leads to increased mitophagy in the human brain, which was also detected using TBI models in male rats. Knockdown of CL synthase, responsible for de novo synthesis of CL, or phospholipid scramblase-3, responsible for CL translocation to the outer mitochondrial membrane, significantly decreased TBI-induced mitophagy. Inhibition of mitochondrial clearance by 3-methyladenine, mdivi-1, or phospholipid scramblase-3 knockdown after TBI led to a worse outcome, suggesting that mitophagy is beneficial. Together, our findings indicate that TBI-induced mitophagy is an endogenous neuroprotective process that is directed by CL, which marks damaged mitochondria for elimination, thereby limiting neuronal death and behavioral deficits.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) increases energy demands leading to activation of mitochondrial respiration associated with enhanced generation of reactive oxygen species and resultant damage to mitochondria. We demonstrate that the complete elimination of irreparably damaged organelles via mitophagy is activated as an early response to TBI. This response includes translocation of mitochondria phospholipid cardiolipin from the inner membrane to the outer membrane where externalized cardiolipin mediates targeted protein light chain 3-mediated autophagy of damaged mitochondria. Our data on targeting phospholipid scramblase and cardiolipin synthase in genetically manipulated cells and animals strongly support the essential role of cardiolipin externalization mechanisms in the endogenous reparative plasticity of injured brain cells. Furthermore, successful execution and completion of mitophagy is beneficial in the context of preservation of cognitive functions after TBI.
Collapse
Affiliation(s)
- Honglu Chao
- Departments of Neurosurgery and
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | | | - Qiang Zuo
- Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | | | - Mengqing Xiao
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
- University of the Chinese Academy of Sciences, CAS, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | | | | | | | - Huimei Chen
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210029, China
| | | | - Patrick M Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
- University of the Chinese Academy of Sciences, CAS, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing 100022, China
| | | | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Laboratory of Navigational Redox Lipidomics and Department of Human Pathology, IM Sechenov Moscow State Medical University, Moscow 119991, Russian Federation, and
| | - Hülya Bayır
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health,
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Jing Ji
- Departments of Neurosurgery and
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| |
Collapse
|
39
|
Lautz AJ, Morgan RW, Karlsson M, Mavroudis CD, Ko TS, Licht DJ, Nadkarni VM, Berg RA, Sutton RM, Kilbaugh TJ. Hemodynamic-Directed Cardiopulmonary Resuscitation Improves Neurologic Outcomes and Mitochondrial Function in the Heart and Brain. Crit Care Med 2019; 47:e241-e249. [PMID: 30779720 PMCID: PMC6561502 DOI: 10.1097/ccm.0000000000003620] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Less than half of the thousands of children who suffer in-hospital cardiac arrests annually survive, and neurologic injury is common among survivors. Hemodynamic-directed cardiopulmonary resuscitation improves short-term survival, but its impact on longer term survival and mitochondrial respiration-a potential neurotherapeutic target-remains unknown. The primary objectives of this study were to compare rates of 24-hour survival with favorable neurologic outcome after cardiac arrest treated with hemodynamic-directed cardiopulmonary resuscitation versus standard depth-guided cardiopulmonary resuscitation and to compare brain and heart mitochondrial respiration between groups 24 hours after resuscitation. DESIGN Randomized preclinical large animal trial. SETTING A large animal resuscitation laboratory at a large academic children's hospital. SUBJECTS Twenty-eight 4-week-old female piglets (8-11 kg). INTERVENTIONS Twenty-two swine underwent 7 minutes of asphyxia followed by ventricular fibrillation and randomized treatment with either hemodynamic-directed cardiopulmonary resuscitation (n = 10; compression depth titrated to aortic systolic pressure of 90 mm Hg, vasopressors titrated to coronary perfusion pressure ≥ 20 mm Hg) or depth-guided cardiopulmonary resuscitation (n = 12; depth 1/3 chest diameter, epinephrine every 4 min). Six animals (sham group) underwent anesthesia and instrumentation without cardiac arrest. The primary outcomes were favorable neurologic outcome (swine Cerebral Performance Category ≤ 2) and mitochondrial maximal oxidative phosphorylation utilizing substrate for complex I and complex II (OXPHOSCI+CII) in the cerebral cortex and hippocampus. MEASUREMENTS AND MAIN RESULTS Favorable neurologic outcome was more likely with hemodynamic-directed cardiopulmonary resuscitation (7/10) than depth-guided cardiopulmonary resuscitation (1/12; p = 0.006). Hemodynamic-directed cardiopulmonary resuscitation resulted in higher intra-arrest coronary perfusion pressure, aortic pressures, and brain tissue oxygenation. Hemodynamic-directed cardiopulmonary resuscitation resulted in higher OXPHOSCI+CII (pmol oxygen/s × mg/citrate synthase) in the cortex (6.00 ± 0.28 vs 3.88 ± 0.43; p < 0.05) and hippocampus (6.26 ± 0.67 vs 3.55 ± 0.65; p < 0.05) and higher complex I respiration (pmol oxygen/s × mg) in the right (20.62 ± 1.06 vs 15.88 ± 0.81; p < 0.05) and left ventricles (20.14 ± 1.40 vs 14.17 ± 1.53; p < 0.05). CONCLUSIONS In a model of asphyxia-associated pediatric cardiac arrest, hemodynamic-directed cardiopulmonary resuscitation increases rates of 24-hour survival with favorable neurologic outcome, intra-arrest hemodynamics, and cerebral and myocardial mitochondrial respiration.
Collapse
Affiliation(s)
- Andrew J. Lautz
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Anesthesiology and Critical Care Medicine
- Cincinnati Children’s Hospital Medical Center; Division of Critical Care Medicine
| | - Ryan W. Morgan
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Anesthesiology and Critical Care Medicine
| | - Michael Karlsson
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Anesthesiology and Critical Care Medicine
| | - Constantine D. Mavroudis
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Cardiothoracic Surgery
| | - Tiffany S. Ko
- University of Pennsylvania, Department of Bioengineering
| | - Daniel J. Licht
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Pediatrics, Division of Neurology
| | - Vinay M. Nadkarni
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Anesthesiology and Critical Care Medicine
| | - Robert A. Berg
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Anesthesiology and Critical Care Medicine
| | - Robert M. Sutton
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Anesthesiology and Critical Care Medicine
| | - Todd J. Kilbaugh
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine; Department of Anesthesiology and Critical Care Medicine
| |
Collapse
|
40
|
Kinder HA, Baker EW, West FD. The pig as a preclinical traumatic brain injury model: current models, functional outcome measures, and translational detection strategies. Neural Regen Res 2019; 14:413-424. [PMID: 30539807 PMCID: PMC6334610 DOI: 10.4103/1673-5374.245334] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a major contributor of long-term disability and a leading cause of death worldwide. A series of secondary injury cascades can contribute to cell death, tissue loss, and ultimately to the development of functional impairments. However, there are currently no effective therapeutic interventions that improve brain outcomes following TBI. As a result, a number of experimental TBI models have been developed to recapitulate TBI injury mechanisms and to test the efficacy of potential therapeutics. The pig model has recently come to the forefront as the pig brain is closer in size, structure, and composition to the human brain compared to traditional rodent models, making it an ideal large animal model to study TBI pathophysiology and functional outcomes. This review will focus on the shared characteristics between humans and pigs that make them ideal for modeling TBI and will review the three most common pig TBI models-the diffuse axonal injury, the controlled cortical impact, and the fluid percussion models. It will also review current advances in functional outcome assessment measures and other non-invasive, translational TBI detection and measurement tools like biomarker analysis and magnetic resonance imaging. The use of pigs as TBI models and the continued development and improvement of translational assessment modalities have made significant contributions to unraveling the complex cascade of TBI sequela and provide an important means to study potential clinically relevant therapeutic interventions.
Collapse
Affiliation(s)
- Holly A Kinder
- Regenerative Bioscience Center; Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| | - Emily W Baker
- Regenerative Bioscience Center; Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| | - Franklin D West
- Regenerative Bioscience Center; Department of Animal and Dairy Science, University of Georgia, Athens, GA, USA
| |
Collapse
|
41
|
Karlsson M, Pukenas B, Chawla S, Ehinger JK, Plyler R, Stolow M, Gabello M, Hugerth M, Elmér E, Hansson MJ, Margulies S, Kilbaugh T. Neuroprotective Effects of Cyclosporine in a Porcine Pre-Clinical Trial of Focal Traumatic Brain Injury. J Neurotrauma 2018; 36:14-24. [PMID: 29929438 PMCID: PMC6306685 DOI: 10.1089/neu.2018.5706] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial dysfunction is thought to be a hallmark of traumatic brain injury (TBI) and plays a pivotal role in the resulting cellular injury. Cyclophilin D-mediated activation of the mitochondrial permeability transition pore has been suggested to contribute to this secondary injury cascade. Cyclosporine possesses neuroprotective properties that have been attributed to the desensitization of mitochondrial permeability transition pore activation. In vivo animal experiments have demonstrated neuroprotective effects of cyclosporine in more than 20 independent experimental studies in a multitude of different experimental models. However, the majority of these studies have been carried out in rodents. The aim of the present study was to evaluate the efficacy of a novel and cremophor/kolliphor EL-free lipid emulsion formulation of cyclosporine in a translational large animal model of TBI. A mild-to-moderate focal contusion injury was induced in piglets using a controlled cortical impact device. After initial step-wise analyses of pharmacokinetics and comparing with exposure of cyclosporine in clinical TBI trials, a 5-day dosing regimen with continuous intravenous cyclosporine infusion (20 mg/kg/day) was evaluated in a randomized and blinded placebo-controlled setting. Cyclosporine reduced the volume of parenchymal injury by 35%, as well as improved markers of neuronal injury, as measured with magnetic resonance spectroscopic imaging. Further, a consistent trend toward positive improvements in brain metabolism and mitochondrial function was observed in the pericontusional tissue. In this study, we have demonstrated efficacy using a novel cyclosporine formulation in clinically relevant and translatable outcome metrics in a large animal model of focal TBI.
Collapse
Affiliation(s)
- Michael Karlsson
- 1 Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
- 2 Mitochondrial Medicine, Department of Clinical Sciences, Lund University , Lund, Sweden
- 3 Department of Neurosurgery, Rigshospitalet , Copenhagen, Denmark
- 4 NeuroVive Pharmaceutical AB , Lund, Sweden
| | - Bryan Pukenas
- 5 Department of Radiology, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Sanjeev Chawla
- 5 Department of Radiology, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Johannes K Ehinger
- 1 Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
- 2 Mitochondrial Medicine, Department of Clinical Sciences, Lund University , Lund, Sweden
- 4 NeuroVive Pharmaceutical AB , Lund, Sweden
| | - Ross Plyler
- 6 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Madeline Stolow
- 6 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Melissa Gabello
- 1 Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | | | - Eskil Elmér
- 2 Mitochondrial Medicine, Department of Clinical Sciences, Lund University , Lund, Sweden
- 4 NeuroVive Pharmaceutical AB , Lund, Sweden
| | - Magnus J Hansson
- 2 Mitochondrial Medicine, Department of Clinical Sciences, Lund University , Lund, Sweden
- 4 NeuroVive Pharmaceutical AB , Lund, Sweden
| | - Susan Margulies
- 6 Department of Bioengineering, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Todd Kilbaugh
- 1 Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
42
|
Gonçalves DF, de Carvalho NR, Leite MB, Courtes AA, Hartmann DD, Stefanello ST, da Silva IK, Franco JL, Soares FA, Dalla Corte CL. Caffeine and acetaminophen association: Effects on mitochondrial bioenergetics. Life Sci 2018; 193:234-241. [DOI: 10.1016/j.lfs.2017.10.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 10/18/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022]
|
43
|
Huang L, Zhang K, Guo Y, Huang F, Yang K, Chen L, Huang K, Zhang F, Long Q, Yang Q. Honokiol protects against doxorubicin cardiotoxicity via improving mitochondrial function in mouse hearts. Sci Rep 2017; 7:11989. [PMID: 28931882 PMCID: PMC5607346 DOI: 10.1038/s41598-017-12095-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/04/2017] [Indexed: 12/15/2022] Open
Abstract
Honokiol is a key component of a medicinal herb, Magnolia bark. Honokiol possesses potential pharmacological benefits for many disease conditions, especially cancer. Recent studies demonstrate that Honokiol exerts beneficial effects on cardiac hypertrophy and doxorubicin (Dox)-cardiotoxicity via deacetylation of mitochondrial proteins. However, the effects and mechanisms of Honokiol on cardiac mitochondrial respiration remain unclear. In the present study, we investigate the effect of Honokiol on cardiac mitochondrial respiration in mice subjected to Dox treatment. Oxygen consumption in freshly isolated mitochondria from mice treated with Honokiol showed enhanced mitochondrial respiration. The Dox-induced impairment of mitochondrial respiration was less pronounced in honokiol-treated than control mice. Furthermore, Luciferase reporter assay reveals that Honokiol modestly increased PPARγ transcriptional activities in cultured embryonic rat cardiomyocytes (H9c2). Honokiol upregulated the expression of PPARγ in the mouse heart. Honokiol repressed cardiac inflammatory responses and oxidative stress in mice subjected to Dox treatment. As a result, Honokiol alleviated Dox-cardiotoxicity with improved cardiac function and reduced cardiomyocyte apoptosis. We conclude that Honokiol protects the heart from Dox-cardiotoxicity via improving mitochondrial function by not only repressing mitochondrial protein acetylation but also enhancing PPARγ activity in the heart. This study further supports Honokiol as a promising therapy for cancer patients receiving Dox treatment.
Collapse
Affiliation(s)
- Lizhen Huang
- School of Basic Medicine, Research Center of Integrative Medicine, Guangzhou University of Chinese Medicine, 230 Guangzhou University City Outer Ring Road, Guangzhou, 510006, China.,Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Kailiang Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Yingying Guo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Fengyuan Huang
- Department of Nutrition Sciences, University of Alabama at Birmingham, 1675 Univ Blvd, Birmingham, AL, 35205, USA
| | - Kevin Yang
- Department of Nutrition Sciences, University of Alabama at Birmingham, 1675 Univ Blvd, Birmingham, AL, 35205, USA
| | - Long Chen
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
| | - Kai Huang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
| | - Fengxue Zhang
- School of Basic Medicine, Research Center of Integrative Medicine, Guangzhou University of Chinese Medicine, 230 Guangzhou University City Outer Ring Road, Guangzhou, 510006, China
| | - Qinqiang Long
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| | - Qinglin Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China. .,Department of Nutrition Sciences, University of Alabama at Birmingham, 1675 Univ Blvd, Birmingham, AL, 35205, USA.
| |
Collapse
|
44
|
Jackson EK, Kotermanski SE, Menshikova EV, Dubey RK, Jackson TC, Kochanek PM. Adenosine production by brain cells. J Neurochem 2017; 141:676-693. [PMID: 28294336 DOI: 10.1111/jnc.14018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 02/06/2023]
Abstract
The early release of adenosine following traumatic brain injury (TBI) suppresses seizures and brain inflammation; thus, it is important to elucidate the cellular sources of adenosine following injurious stimuli triggered by TBI so that therapeutics for enhancing the early adenosine-release response can be optimized. Using mass spectrometry with 13 C-labeled standards, we investigated in cultured rat neurons, astrocytes, and microglia the effects of oxygen-glucose deprivation (OGD; models energy failure), H2 O2 (produces oxidative stress), and glutamate (induces excitotoxicity) on intracellular and extracellular levels of 5'-AMP (adenosine precursor), adenosine, and inosine and hypoxanthine (adenosine metabolites). In neurons, OGD triggered increases in intracellular 5'-AMP (2.8-fold), adenosine (2.6-fold), inosine (2.2-fold), and hypoxanthine (5.3-fold) and extracellular 5'-AMP (2.2-fold), adenosine (2.4-fold), and hypoxanthine (2.5-fold). In neurons, H2 O2 did not affect intracellular or extracellular purines; yet, glutamate increased intracellular adenosine, inosine, and hypoxanthine (1.7-fold, 1.7-fold, and 1.6-fold, respectively) and extracellular adenosine, inosine, and hypoxanthine (2.9-fold, 2.1-fold, and 1.6-fold, respectively). In astrocytes, neither H2 O2 nor glutamate affected intracellular or extracellular purines, and OGD only slightly increased intracellular and extracellular hypoxanthine. Microglia were unresponsive to OGD and glutamate, but were remarkably responsive to H2 O2 , which increased intracellular 5'-AMP (1.6-fold), adenosine (1.6-fold), inosine (2.1-fold), and hypoxanthine (1.6-fold) and extracellular 5'-AMP (5.9-fold), adenosine (4.0-fold), inosine (4.3-fold), and hypoxanthine (1.9-fold). CONCLUSION Under these particular experimental conditions, cultured neurons are the main contributors to adenosine production/release in response to OGD and glutamate, whereas cultured microglia are the main contributors upon oxidative stress. Developing therapeutics that recruit astrocytes to produce/release adenosine could have beneficial effects in TBI.
Collapse
Affiliation(s)
- Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shawn E Kotermanski
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elizabeth V Menshikova
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Raghvendra K Dubey
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Reproductive Endocrinology, University Hospital Zurich and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Travis C Jackson
- Department of Critical Care Medicine and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine and the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Li X, Wang H, Gao Y, Li L, Tang C, Wen G, Zhou Y, Zhou M, Mao L, Fan Y. Protective Effects of Quercetin on Mitochondrial Biogenesis in Experimental Traumatic Brain Injury via the Nrf2 Signaling Pathway. PLoS One 2016; 11:e0164237. [PMID: 27780244 PMCID: PMC5079551 DOI: 10.1371/journal.pone.0164237] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/21/2016] [Indexed: 01/17/2023] Open
Abstract
The present investigation was carried out to elucidate a possible molecular mechanism related to the protective effect of quercetin administration against oxidative stress on various mitochondrial respiratory complex subunits with special emphasis on the role of nuclear factor erythroid 2-related factor 2 (Nrf2) in mitochondrial biogenesis. Recently, quercetin has been proved to have a protective effect against mitochondria damage after traumatic brain injury (TBI). However, its precise role and underlying mechanisms in traumatic brain injury are not yet fully understood. The aim of the present study was to investigate the effect of quercetin on the potential mechanism of these effects in a weight-drop model of TBI in male mice that were treated with quercetin or vehicle via intraperitoneal injection administrated 30 min after TBI. In this experiment, ICR mice were divided into four groups: A sham group, TBI group, TBI + vehicle group, and TBI + quercetin group. Brain samples were collected 24 h later for analysis. Quercetin treatment resulted in an upregulation of Nrf2 expression and cytochrome c, malondialdehyde (MDA) and superoxide dismutase (SOD) levels were restored by quercetin treatment. Quercetin markedly promoted the translocation of Nrf2 protein from the cytoplasm to the nucleus. These observations suggest that quercetin improves mitochondrial function in TBI models, possibly by activating the Nrf2 pathway.
Collapse
Affiliation(s)
- Xiang Li
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Yongyue Gao
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Liwen Li
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Chao Tang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Guodao Wen
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Yuan Zhou
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Mengliang Zhou
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Lei Mao
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| | - Youwu Fan
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, No. 305 East Zhongshan Road, Nanjing, Jiangsu Province, 210002, China
| |
Collapse
|
46
|
Wong VS, Langley B. Epigenetic changes following traumatic brain injury and their implications for outcome, recovery and therapy. Neurosci Lett 2016; 625:26-33. [PMID: 27155457 PMCID: PMC4915732 DOI: 10.1016/j.neulet.2016.04.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/03/2016] [Accepted: 04/05/2016] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) contributes to nearly a third of all injury-related deaths in the United States. For survivors of TBI, depending on severity, patients can be left with devastating neurological disabilities that include impaired cognition or memory, movement, sensation, or emotional function. Despite the efforts to identify novel therapeutics, the only strategy to combat TBI is risk reduction (helmets, seatbelts, removal of fall hazards, etc.). Enormous heterogeneity exists within TBI, and it depends on the severity, the location, and whether the injury was focal or diffuse. Evidence from recent studies support the involvement of epigenetic mechanisms such as DNA methylation, chromatin post-translational modification, and miRNA regulation of gene expression in the post-injured brain. In this review, we discuss studies that have assessed epigenetic changes and mechanisms following TBI, how epigenetic changes might not only be limited to the nucleus but also impact the mitochondria, and the implications of these changes with regard to TBI recovery.
Collapse
Affiliation(s)
- Victor S Wong
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, United States
| | - Brett Langley
- Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, United States; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, 525 E. 68th Street, New York, NY 10065, United States.
| |
Collapse
|
47
|
Ferguson MA, Sutton RM, Karlsson M, Sjövall F, Becker LB, Berg RA, Margulies SS, Kilbaugh TJ. Increased platelet mitochondrial respiration after cardiac arrest and resuscitation as a potential peripheral biosignature of cerebral bioenergetic dysfunction. J Bioenerg Biomembr 2016; 48:269-79. [PMID: 27020568 DOI: 10.1007/s10863-016-9657-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/15/2016] [Indexed: 02/02/2023]
Abstract
UNLABELLED Cardiac arrest (CA) results in a sepsis-like syndrome with activation of the innate immune system and increased mitochondrial bioenergetics. OBJECTIVE To determine if platelet mitochondrial respiration increases following CA in a porcine pediatric model of asphyxia-associated ventricular fibrillation (VF) CA, and if this readily obtained biomarker is associated with decreased brain mitochondrial respiration. CA protocol: 7 min of asphyxia, followed by VF, protocolized titration of compression depth to systolic blood pressure of 90 mmHg and vasopressor administration to a coronary perfusion pressure greater than 20 mmHg. PRIMARY OUTCOME platelet integrated mitochondrial electron transport system (ETS) function evaluated pre- and post-CA/ROSC four hours after return of spontaneous circulation (ROSC). Secondary outcome: correlation of platelet mitochondrial bioenergetics to cerebral bioenergetic function. Platelet maximal oxidative phosphorylation (OXPHOSCI+CII), P < 0.02, and maximal respiratory capacity (ETSCI+CII), P < 0.04, were both significantly increased compared to pre-arrest values. This was primarily due to a significant increase in succinate-supported respiration through Complex II (OXPHOSCII, P < 0.02 and ETSCII, P < 0.03). Higher respiration was not due to uncoupling, as the LEAKCI + CII respiration (mitochondrial respiration independent of ATP-production) was unchanged after CA/ROSC. Larger increases in platelet mitochondrial respiratory control ratio (RCR) compared to pre-CA RCR were significantly correlated with lower RCRs in the cortex (P < 0.03) and hippocampus (P < 0.04) compared to sham respiration. Platelet mitochondrial respiration is significantly increased four hours after ROSC. Future studies will identify mechanistic relationships between this serum biomarker and altered cerebral bioenergetics function following cardiac arrest.
Collapse
Affiliation(s)
- Michael A Ferguson
- Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 34th & Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Robert M Sutton
- Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 34th & Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Michael Karlsson
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84, Lund, Sweden
| | - Fredrik Sjövall
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84, Lund, Sweden
| | - Lance B Becker
- Department of Emergency Medicine, Perelman School of Medicine at the University of Pennsylvania, The Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Robert A Berg
- Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 34th & Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Susan S Margulies
- School of Engineering and Applied Science, Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Todd J Kilbaugh
- Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 34th & Civic Center Blvd., Philadelphia, PA, 19104, USA.
| |
Collapse
|
48
|
Kilbaugh TJ, Karlsson M, Duhaime AC, Hansson MJ, Elmer E, Margulies SS. Mitochondrial response in a toddler-aged swine model following diffuse non-impact traumatic brain injury. Mitochondrion 2016; 26:19-25. [PMID: 26549476 PMCID: PMC4752861 DOI: 10.1016/j.mito.2015.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 01/19/2023]
Abstract
Traumatic brain injury (TBI) is an important health problem, and a leading cause of death in children worldwide. Mitochondrial dysfunction is a critical component of the secondary TBI cascades. Mitochondrial response in the pediatric brain has limited investigation, despite evidence that the developing brain's response differs from that of the adult, especially in diffuse non-impact TBI. We performed a detailed evaluation of mitochondrial bioenergetics using high-resolution respirometry in a swine model of diffuse TBI (rapid non-impact rotational injury: RNR), and examined the cortex and hippocampus. A substrate-uncoupler-inhibitor-titration protocol examined the role of the individual complexes as well as the uncoupled maximal respiration. Respiration per mg of tissue was also related to citrate synthase activity (CS) as an attempt to control for variability in mitochondrial content following injury. Diffuse RNR stimulated increased complex II-driven respiration relative to mitochondrial content in the hippocampus compared to shams. LEAK (State 4o) respiration increased in both regions, with decreased respiratory ratios of convergent oxidative phosphorylation through complex I and II, compared to sham animals, indicating uncoupling of oxidative phosphorylation at 24h. The study suggests that proportionately, complex I contribution to convergent mitochondrial respiration was reduced in the hippocampus after RNR, with a simultaneous increase in complex-II driven respiration. Mitochondrial respiration 24h after diffuse TBI varies by location within the brain. We concluded that significant uncoupling of oxidative phosphorylation and alterations in convergent respiration through complex I- and complex II-driven respiration reveals therapeutic opportunities for the injured at-risk pediatric brain.
Collapse
Affiliation(s)
- Todd J Kilbaugh
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, 3401 Civic Center Blvd., Philadelphia, PA 19104, USA.
| | - Michael Karlsson
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84 Lund, Sweden.
| | - Ann-Christine Duhaime
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, PA 19104, USA.
| | - Magnus J Hansson
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84 Lund, Sweden.
| | - Eskil Elmer
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84 Lund, Sweden.
| | - Susan S Margulies
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, Boston, MA 02114, USA.
| |
Collapse
|
49
|
Kilbaugh TJ, Sutton RM, Karlsson M, Hansson MJ, Naim MY, Morgan RW, Bratinov G, Lampe JW, Nadkarni VM, Becker LB, Margulies SS, Berg RA. Persistently Altered Brain Mitochondrial Bioenergetics After Apparently Successful Resuscitation From Cardiac Arrest. J Am Heart Assoc 2015; 4:e002232. [PMID: 26370446 PMCID: PMC4599507 DOI: 10.1161/jaha.115.002232] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Although advances in cardiopulmonary resuscitation have improved survival from cardiac arrest (CA), neurologic injury persists and impaired mitochondrial bioenergetics may be critical for targeted neuroresuscitation. The authors sought to determine if excellent cardiopulmonary resuscitation and postresuscitation care and good traditional survival rates result in persistently disordered cerebral mitochondrial bioenergetics in a porcine pediatric model of asphyxia-associated ventricular fibrillation CA. METHODS AND RESULTS After 7 minutes of asphyxia, followed by ventricular fibrillation, 5 female 1-month-old swine (4 sham) received blood pressure-targeted care: titration of compression depth to systolic blood pressure of 90 mm Hg and vasopressor administration to a coronary perfusion pressure >20 mm Hg. All animals received protocol-based vasopressor support after return of spontaneous circulation for 4 hours before they were killed. The primary outcome was integrated mitochondrial electron transport system (ETS) function. CA animals displayed significantly decreased maximal, coupled oxidative phosphorylating respiration (OXPHOSCI + CII) in cortex (P<0.02) and hippocampus (P<0.02), as well as decreased phosphorylation and coupling efficiency (cortex, P<0.05; hippocampus, P<0.05). Complex I- and complex II-driven respiration were both significantly decreased after CA (cortex: OXPHOSCI P<0.01, ETSCII P<0.05; hippocampus: OXPHOSCI P<0.03, ETSCII P<0.01). In the hippocampus, there was a significant decrease in maximal uncoupled, nonphosphorylating respiration (ETSCI + CII), as well as a 30% reduction in citrate synthase activity (P<0.04). CONCLUSIONS Mitochondria in both the cortex and hippocampus displayed significant alterations in respiratory function after CA despite excellent cardiopulmonary resuscitation and postresuscitation care in asphyxia-associated ventricular fibrillation CA. Analysis of integrated ETS function identifies mitochondrial bioenergetic failure as a target for goal-directed neuroresuscitation after CA. IACUC Protocol: IAC 13-001023.
Collapse
Affiliation(s)
- Todd J Kilbaugh
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (T.J.K., R.M.S., M.Y.N., R.W.M., G.B., V.M.N., R.A.B.)
| | - Robert M Sutton
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (T.J.K., R.M.S., M.Y.N., R.W.M., G.B., V.M.N., R.A.B.)
| | - Michael Karlsson
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden (M.K., M.J.H.)
| | - Magnus J Hansson
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden (M.K., M.J.H.)
| | - Maryam Y Naim
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (T.J.K., R.M.S., M.Y.N., R.W.M., G.B., V.M.N., R.A.B.)
| | - Ryan W Morgan
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (T.J.K., R.M.S., M.Y.N., R.W.M., G.B., V.M.N., R.A.B.)
| | - George Bratinov
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (T.J.K., R.M.S., M.Y.N., R.W.M., G.B., V.M.N., R.A.B.)
| | - Joshua W Lampe
- Department of Emergency Medicine, The Hospital of the University of Pennsylvania, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (J.W.L., L.B.B.)
| | - Vinay M Nadkarni
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (T.J.K., R.M.S., M.Y.N., R.W.M., G.B., V.M.N., R.A.B.)
| | - Lance B Becker
- Department of Emergency Medicine, The Hospital of the University of Pennsylvania, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (J.W.L., L.B.B.)
| | - Susan S Margulies
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA (S.S.M.)
| | - Robert A Berg
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA (T.J.K., R.M.S., M.Y.N., R.W.M., G.B., V.M.N., R.A.B.)
| |
Collapse
|